praise from the experts - bayanbox.irbayanbox.ir/.../pharmaceutical-statistics-using-sas.pdf ·...

460

Upload: phungcong

Post on 13-May-2018

256 views

Category:

Documents


7 download

TRANSCRIPT

Page 1: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 2: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications of cutting-edge statistical techniques using cutting-edge software tools provided by SAS. The theory is presented in down-to-earth ways, with copious examples, for simple understanding. For pharmaceutical statisticians, connections with appropriate guidance documents are made; the connections between the document and the data analysis techniques make ‘standard practice’ easy to implement. In addition, the included references make it easy to find these guidance documents that are often obscure.

“Specialized procedures, such as easy calculation of the power of nonparametric and survival analysis tests, are made transparent, and this should be a delight to the statistician working in the pharmaceutical industry, who typically spends long hours on such calculations. However, non-pharmaceutical statisticians and scientists will also appreciate the treatment of problems that are more generally common, such as how to handle dropouts and missing values, assessing reliability and validity of psychometric scales, and decision theory in experimental design. I heartily recommend this book to all.”

Peter H. Westfall Professor of Statistics

Texas Tech University

“The book is well written by people well known in the pharmaceutical industry. The selected topics are comprehensive and relevant. Explanations of the statistical theory are concise, and the solutions are up-to-date. It would be particularly useful for isolated statisticians who work for companies without senior colleagues.”

Frank Shen Executive Director Global Biometric Sciences Bristol-Myers Squibb Co.

“This book covers an impressive range of topics in clinical and non-clinical statistics. Adding the fact that all the datasets and SAS code discussed in the book are available on the SAS Web site, this book will be a very useful resource for statisticians in the pharmaceutical industry.”

Professor Byron Jones Senior Director Pfizer Global Research and Development, UK

Page 3: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

“The first thing that catches one’s attention about this very interesting book is its breadth of coverage of statistical methods applied to pharmaceutical drug development. Starting with drug discovery, moving through pre-clinical and non-clinical applications, and concluding with many relevant topics in clinical development, the book provides a comprehensive reference to practitioners involved in, or just interested to learn about, any stage of drug development. “There is a good balance between well-established and novel material, making the book attractive to both newcomers to the field and experienced pharmaceutical statisticians. The inclusion of examples from real studies, with SAS code implementing the corresponding methods, in every chapter but the introduction, is particularly useful to those interested in applying the methods in practice, and who certainly will be the majority of the readers. Overall, an excellent addition to the SAS Press collection.”

José Pinheiro Director of Biostatistics Novartis Pharmaceuticals

“This is a very well-written, state-of-the-art book that covers a wide range of statistical issues through all phases of drug development. It represents a well-organized and thorough exploration of many of the important aspects of statistics as used in the pharmaceutical industry. The book is packed with useful examples and worked exercises using SAS. The underlying statistical methodology that justifies the methods used is clearly presented. “The authors are clearly expert and have done an excellent job of linking the various statistical applications to research problems in the pharmaceutical industry. Many areas are covered including model building, nonparametric methods, pharmacokinetic analysis, sample size estimation, dose-ranging studies, and decision analysis. This book should serve as an excellent resource for statisticians and scientists engaged in pharmaceutical research or anyone who wishes to learn about the role of the statistician in the pharmaceutical industry.”

Barry R. Davis Professor of Biomathematics University of Texas

Page 4: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Pharmaceutical Statistics

Edited byAlex Dmitrienko

Christy Chuang-SteinRalph D’Agostino

Using SAS®

A Practical Guide

Page 5: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

The correct bibliographic citation for this manual is as follows: Dmitrienko, Alex, Christy Chuang-Stein, and Ralph D’Agostino. 2007. Pharmaceutical Statistics Using SAS®: A Practical Guide. Cary, NC: SAS Institute Inc.

Pharmaceutical Statistics Using SAS®: A Practical Guide

Copyright © 2007, SAS Institute Inc., Cary, NC, USA

ISBN: 978-1-59047-886-8

All rights reserved. Produced in the United States of America.

For a hard-copy book: No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic, mechanical, photocopying, or otherwise, without the prior written permission of the publisher, SAS Institute Inc.

For a Web download or e-book: Your use of this publication shall be governed by the terms established by the vendor at the time you acquire this publication.

U.S. Government Restricted Rights Notice: Use, duplication, or disclosure of this software and related documentation by the U.S. government is subject to the Agreement with SAS Institute and the restrictions set forth in FAR 52.227-19, Commercial Computer Software-Restricted Rights (June 1987).

SAS Institute Inc., SAS Campus Drive, Cary, North Carolina 27513.

1st printing, February 2007

SAS® Publishing provides a complete selection of books and electronic products to help customers use SAS software to its fullest potential. For more information about our e-books, e-learning products, CDs, and hard-copy books, visit the SAS Publishing Web site at support.sas.com/pubs or call 1-800-727-3228.

SAS® and all other SAS Institute Inc. product or service names are registered trademarks or trademarks of SAS Institute Inc. in the USA and other countries. ® indicates USA registration.

Other brand and product names are registered trademarks or trademarks of their respective companies.

Page 6: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Contents

1 Statistics in Drug Development 1By Christy Chuang-Stein and Ralph D’Agostino

1.1 Introduction 11.2 Statistical Support to Non-Clinical Activities 21.3 Statistical Support to Clinical Testing 31.4 Battling a High Phase III Failure Rate 41.5 Do Statisticians Count? 51.6 Emerging Opportunities 51.7 Summary 6

References 6

2 Modern Classification Methods for Drug Discovery 7By Kjell Johnson and William Rayens

2.1 Introduction 72.2 Motivating Example 92.3 Boosting 102.4 Model Building 272.5 Partial Least Squares for Discrimination 332.6 Summary 42

References 42

3 Model Building Techniques in Drug Discovery 45By Kimberly Crimin and Thomas Vidmar

3.1 Introduction 453.2 Example: Solubility Data 463.3 Training and Test Set Selection 473.4 Variable Selection 513.5 Statistical Procedures for Model Building 583.6 Determining When a New Observation Is Not in a Training Set 613.7 Using SAS Enterprise Miner 633.8 Summary 67

References 67

Page 7: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

iv Pharmaceutical Statistics Using SAS: A Practical Guide

4 Statistical Considerations in Analytical Method Validation 69By Bruno Boulanger, Viswanath Devanaryan, Walthere Dewe,and Wendell Smith

4.1 Introduction 694.2 Validation Criteria 734.3 Response Function or Calibration Curve 744.4 Linearity 834.5 Accuracy and Precision 854.6 Decision Rule 884.7 Limits of Quantification and Range of the Assay 924.8 Limit of Detection 934.9 Summary 93

4.10 Terminology 94References 94

5 Some Statistical Considerations in Nonclinical Safety Assessment 97By Wherly Hoffman, Cindy Lee, Alan Chiang, Kevin Guo, and Daniel Ness

5.1 Overview of Nonclinical Safety Assessment 975.2 Key Statistical Aspects of Toxicology Studies 985.3 Randomization in Toxicology Studies 995.4 Power Evaluation in a Two-Factor Model for QT Interval 1025.5 Statistical Analysis of a One-Factor Design with Repeated Measures 1065.6 Summary 113

Acknowledgments 115References 115

6 Nonparametric Methods in Pharmaceutical Statistics 117By Paul Juneau

6.1 Introduction 1176.2 Two Independent Samples Setting 1186.3 The One-Way Layout 1296.4 Power Determination in a Purely Nonparametric Sense 144

Acknowledgments 149References 149

7 Optimal Design of Experiments in Pharmaceutical Applications 151By Valerii Fedorov, Robert Gagnon, Sergei Leonov, and Yuehui Wu

7.1 Optimal Design Problem 1527.2 Quantal Dose-Response Models 1597.3 Nonlinear Regression Models with a Continuous Response 1657.4 Regression Models with Unknown Parameters in the Variance Function 1697.5 Models with a Bounded Response (Beta Models) 1727.6 Models with a Bounded Response (Logit Link) 1767.7 Bivariate Probit Models for Correlated Binary Responses 1817.8 Pharmacokinetic Models with Multiple Measurements per Patient 184

Page 8: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Pharmaceutical Statistics Using SAS: A Practical Guide v

7.9 Models with Cost Constraints 1907.10 Summary 192

References 193

8 Analysis of Human Pharmacokinetic Data 197By Scott Patterson and Brian Smith

8.1 Introduction 1978.2 Bioequivalence Testing 1998.3 Assessing Dose Linearity 2048.4 Summary 209

References 209

9 Allocation in Randomized Clinical Trials 213By Olga Kuznetsova and Anastasia Ivanova

9.1 Introduction 2139.2 Permuted Block Randomization 2149.3 Variations of Permuted Block Randomization 2179.4 Allocations Balanced on Baseline Covariates 2289.5 Summary 233

Acknowledgments 233References 233

10 Sample-Size Analysis for Traditional Hypothesis Testing:Concepts and Issues 237By Ralph G. O’Brien and John Castelloe10.1 Introduction 23810.2 Research Question 1: Does “QCA” Decrease Mortality in Children with

Severe Malaria? 24010.3 p-Values, α, β and Power 24110.4 A Classical Power Analysis 24310.5 Beyond α and β: Crucial Type I and Type II Error Rates 24910.6 Research Question 1, Continued: Crucial Error Rates for Mortality Analysis 25110.7 Research Question 2: Does “QCA” Affect the “Elysemine : Elysemate”

Ratios (EER)? 25310.8 Crucial Error Rates When the Null Hypothesis Is Likely to Be True 26210.9 Table of Crucial Error Rates 263

10.10 Summary 263Acknowledgments 264References 264

Appendix A Guidelines for “Statistical Considerations” Sections 264Appendix B SAS Macro Code to Automate the Programming 265

11 Design and Analysis of Dose-Ranging Clinical Studies 273By Alex Dmitrienko, Kathleen Fritsch, Jason Hsu, and Stephen Ruberg11.1 Introduction 27311.2 Design Considerations 277

Page 9: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

vi Pharmaceutical Statistics Using SAS: A Practical Guide

11.3 Detection of Dose-Response Trends 28011.4 Regression Modeling 28911.5 Dose-Finding Procedures 29411.6 Summary 309

References 310

12 Analysis of Incomplete Data 313By Geert Molenberghs, Caroline Beunckens, Herbert Thijs, Ivy Jansen,Geert Verbeke, Michael Kenward, and Kristel Van Steen12.1 Introduction 31412.2 Case Studies 31612.3 Data Setting and Modeling Framework 31812.4 Simple Methods and MCAR 31912.5 MAR Methods 32012.6 Categorical Data 32212.7 MNAR Modeling 34012.8 Sensitivity Analysis 34712.9 Summary 356

References 356

13 Reliability and Validity: Assessing the Psychometric Properties ofRating Scales 361By Douglas Faries and Ilker Yalcin13.1 Introduction 36113.2 Reliability 36213.3 Validity and Other Topics 37613.4 Summary 382

References 383

14 Decision Analysis in Drug Development 385By Carl-Fredrik Burman, Andy Grieve, and Stephen Senn14.1 Introduction 38514.2 Introductory Example: Stop or Go? 38614.3 The Structure of a Decision Analysis 39214.4 The Go/No Go Problem Revisited 39414.5 Optimal Sample Size 39714.6 Sequential Designs in Clinical Trials 40614.7 Selection of an Optimal Dose 41214.8 Project Prioritization 42114.9 Summary 426

Acknowledgments 426References 426

Index 429

Page 10: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Preface

IntroductionThe past decades have witnessed significant developments of biostatistical methodologyapplied to all areas of pharmaceutical drug development. These applications range fromdrug discovery to animal studies to Phase III trials. The use of statistical methods helpsoptimize a variety of drug development processes and ultimately helps ensure that newchemical entities are pure and stable, new therapies are safe and efficacious.

For the most part, new developments in biostatistical theory and applications arescattered across numerous research papers or books on specialized topics and rarely appearunder the same cover. The objective of this book is to offer a broad coverage ofbiostatistical methodology used in drug development and practical problems facing today’sdrug developers.

Each chapter of the book features a discussion of methodological issues, traditional andrecently developed approaches to data analysis, practical advice from subject matterexperts, and review of relevant regulatory guidelines. The book is aimed at practitionersand therefore does not place much emphasis on technical details. It shows how toimplement the algorithms presented in the various chapters by using built-in SASprocedures or custom SAS macros written by the authors. In order to help readers betterunderstand the underlying concepts and facilitate application of the introducedbiostatistical methods, the methods are illustrated with a large number of case studiesfrom actual pre-clinical experiments and clinical trials. Since many of the statistical issuesencountered during late-phase drug development (e.g., survival analysis and interimanalyses) have been covered in other books, this book focuses on statistical methods tosupport research and early drug development activities.

Although the book is written primarily for biostatisticians, it will benefit a broad groupof pharmaceutical researchers, including biologists, chemists, pharmacokineticists, andpharmacologists. Most chapters are self-contained and include a fair amount of high-levelintroductory material. This book will also serve as a useful reference for regulatoryscientists as well as academic researchers and graduate students.

We hope that this book will help close the gap between modern statistical theory andexisting data analysis practices in the pharmaceutical industry. By doing so, we hope thebook will help advance drug discovery and development.

Outline of the BookThe book begins with a review of statistical problems arising in pharmaceutical drugdevelopment (Chapter 1, “Statistics in Drug Development”). This introductory chapter isfollowed by thirteen chapters that deal with statistical approaches used in drug discoveryexperiments, animal toxicology studies, and clinical trials. Since some of the chaptersdiscuss methods that are relevant at multiple stages of drug development (e.g.,nonparametric methods), the chapters in this book are not grouped by the developmentstage. Table helps the reader identify chapters that deal with a particular area inpharmaceutical statistics.

Page 11: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

viii Pharmaceutical Statistics Using SAS: A Practical Guide

Chapter list

Drug Animal ClinicalChapter discovery toxicology trials

Chapter 2, “Modern Classification Methods for •Drug Discovery”

Chapter 3, “Model Building Techniques in •Drug Discovery”

Chapter 4, “Statistical Considerations in Analytical •Method Validation”

Chapter 5, “Some Statistical Considerations in •Nonclinical Safety Assessment”

Chapter 6, “Nonparametric Methods in • •Pharmaceutical Statistics”

Chapter 7, “Optimal Design of Experiments in • •Pharmaceutical Applications”

Chapter 8, “Analysis of Human Pharmacokinetic •Data”

Chapter 9, “Allocation in Randomized Clinical •Trials”

Chapter 10, “Sample-Size Analysis for Traditional •Hypothesis Testing: Concepts and Issues”

Chapter 11, “Design and Analysis of Dose-Ranging •Clinical Studies”

Chapter 12, “Analysis of Incomplete Data” •Chapter 13, “Reliability and Validity: Assessing the •

Psychometric Properties of Rating Scales”Chapter 14, “Decision Analysis in Drug •

Development”

List of ContributorsThis book represents a collaborative effort with contributions from 42 scientists whosenames are listed below.

Caroline Beunckens, Universiteit HasseltBruno Boulanger, Eli LillyCarl-Fredrik Burman, AstraZenecaJohn Castelloe, SAS InstituteAlan Chiang, Eli LillyChristy Chuang-Stein, PfizerKimberly Crimin, WyethRalph D’Agostino, Boston UniversityViswanath Devanaryan, MerckWalthere Dewe, Eli LillyAlex Dmitrienko, Eli LillyDouglas Faries, Eli LillyValerii Fedorov, GlaxoSmithKlineKathleen Fritsch, Food and Drug AdministrationRobert Gagnon, GlaxoSmithKlineAndy Grieve, PfizerKevin Guo, Eli LillyWherly Hoffman, Eli Lilly

Page 12: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Pharmaceutical Statistics Using SAS: A Practical Guide ix

Jason Hsu, The Ohio State UniversityAnastasia Ivanova, University of North CarolinaIvy Jansen, Universiteit HasseltKjell Johnson, PfizerPaul Juneau, PfizerMichael Kenward, London School of Hygiene and Tropical MedicineOlga Kuznetsova, MerckCindy Lee, Eli LillySergei Leonov, GlaxoSmithKlineGeert Molenberghs, Universiteit HasseltDaniel Ness, Eli LillyRalph O’Brien, Cleveland ClinicScott Patterson, GlaxoSmithKlineWilliam Rayens, University of KentuckyStephen Ruberg, Eli LillyStephen Senn, University of GlasgowBrian Smith, AmgenWendell Smith, B2S ConsultingHerbert Thijs, Universiteit HasseltKristel Van Steen, Universitiet GentGeert Verbeke, Katholieke Universiteit LeuvenThomas Vidmar, PfizerYuehui Wu, GlaxoSmithKlineIlker Yalcin, Eli Lilly

FeedbackWe encourage you to submit your comments, share your thoughts, and suggest topics thatwe can consider in other editions of this book on the Biopharmaceutical Network’s website at

http://www.biopharmnet.com/books

We value your opinions and look forward to hearing from you.

AcknowledgmentsWe would like to thank the reviewers, including Dr. Ilya Lipkovich (Eli Lilly andCompany) and Professor Peter Westfall (Texas Tech University), who have provided acareful review of selected chapters and valuable comments.

Alex Dmitrienko wants to thank Drs. Walter Offen, Gregory Sides, and Gregory Enasfor their advice and encouragement throughout the years.

Christy Chuang Stein wishes to thank Drs. Bruce Stein and Mohan Beltangady for theirsupport and encouragement.

Last, but not least, we would like to thank Donna Faircloth, acquisitions editor at SASPress, for all the work and time she has invested in this book publishing project.

Alex Dmitrienko, Global Statistical Sciences, Eli Lilly and Company, USA.Christy Chuang-Stein, Midwest Statistics, Pfizer, USA.Ralph D’Agostino, Department of Mathematics and Statistics, Boston University, USA.

Page 13: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

66

Page 14: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 1

Statistics in DrugDevelopment

Christy Chuang-SteinRalph D’Agostino

1.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1

1.2 Statistical Support to Non-Clinical Activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2

1.3 Statistical Support to Clinical Testing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .3

1.4 Battling a High Phase III Failure Rate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4

1.5 Do Statisticians Count? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5

1.6 Emerging Opportunities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5

1.7 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6

1.1 IntroductionIn the past 50 years, the value of medicine has been clearly demonstrated by a longer lifeexpectancy, a lower infant mortality rate, and the higher quality of life many of our seniorcitizens have been enjoying. Since the introduction of stomach-acid-blocking H2 antagonistdrugs in the late 70’s, the number of surgeries to treat ulcer has been greatly reduced.Childhood vaccination has literally wiped out diphtheria, whooping cough, measles, andpolio in the U.S. Deaths from heart disease have been cut by more than half since 1950 andcontinue to decline. Even though we still face great challenges in combating cancer, greatstrides have been made in treating childhood leukemia. Early detection has led tosuccessful treatment of some types of cancer such as breast cancer. Treatments forschizophrenia and bipolar disorder have allowed many patients to live almost normal lives.A report (2006) on the value of medicine can be found at the Pharmaceutical Research andManufacturers of America (PhRMA) website.

The use of statistics to support discovery and testing of new medicines has grownrapidly since the Kefauver-Harris Amendments, which became effective in 1962. TheKefauver-Harris Amendments required drug sponsors to prove a product’s safety andefficacy in controlled clinical trials in order to market the product. Since the Amendments,the number of statisticians working in the pharmaceutical industry has greatly increased.This increase took another jump when the manufacturing process came under closescrutiny. As we move into the 21st century, the lure and the promise of genomics and

Christy Chuang-Stein is Site Head, Midwest Statistics, Pfizer, USA. Ralph D’Agostino is Professor, Department of Math-ematics and Statistics, Boston University, USA.

Page 15: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

2 Pharmaceutical Statistics Using SAS: A Practical Guide

proteomics will further intensify scientists’ reliance on statistics. The need to enhance ouroverall knowledge about diseases, the need to insert more points into the decision-makingprocess, and the need to bring economics into development strategy considerations willundoubtedly present new opportunities for statisticians.

Even in the face of new opportunities, there are many well-established roles forstatisticians in the pharmaceutical industry. The term “well-established” is a relative termsince new roles will become more established over time. For example, trial simulation andmodeling, viewed as new advancements a decade ago, have now become common practiceto help design better trials across the pharmaceutical industry.

Concerned that the current medical product development path may have becomeincreasingly challenging, inefficient, and costly, the U.S. Food and Drug Administration(FDA) issued a document in March 2004 entitled “Challenge and Opportunity on theCritical Path to New Medical Products”. The document attempts to bridge thetechnological disconnect between discovery and the product development process. Thedisconnect is thought to be largely due to the fact that the pace of development work hasnot kept up with the rapid advances in product discovery. The document addresses threemajor scientific and technical dimensions in the critical path of product development. Thethree dimensions relate to safety assessment, demonstration of a product’s medical utility(benefit or effectiveness), and the product’s industrialization (scaling up). In addition tounderstanding the challenges, establishing the right standards and developing bettertoolkits for each dimension will be key to our ultimate success in overcoming the perceivedstagnation in getting new drugs and biologics to the market. Statisticians, with theirtraining in quantification and logical thinking, can play a major role in the preparation andthe execution of the action plan.

The call for innovation is nothing new for the pharmaceutical industry. The industry asa whole has made great strides in its basic science research in recent years. Cutting edgetechniques are being developed on a daily basis to probe into the biologic origin and geneticconnection of diseases. The research on microarrays and genomics has produced more datathan could be perceived just a few years ago. With the race to unlock the mysteries ofmany diseases and finding cures for them, statistical support needs to be broadened indimensions and increased in depth. Time has never been more right for statisticians towork alongside with their colleagues, being discovery scientists, clinical personnel,manufacturing engineers, or regulatory colleagues. The collaboration should not only helptransform data to knowledge, but also help use knowledge for better risk-based decisions.

In this chapter, we will briefly cover some traditional statistical support to show howstatistics has been used in many aspects of drug development. Our coverage is by no meansexhaustive. It is simply an attempt to illustrate how broad statistical applications havebeen. We will also highlight some areas where a statistician’s contribution will be crucial inmoving forward, in view of the FDA’s Critical Path initiative and the pharmaceuticalindustry’s collective effort to take advantage of the FDA’s call for innovation.

1.2 Statistical Support to Non-Clinical ActivitiesIn an eloquent viewpoint article, Dennis Lendrem (2002) discussed non-clinical statisticalsupport. Traditionally, statistical thinking and approaches are more embraced in areaswhere regulators have issued guidelines. Examples are pre-clinical testing of cardiacliability, carcinogenicity, and stability testing. Recently, Good Manufacturing Practice hasalso become a subject of great regulatory interest. The latter captured public attentionwhen manufacturing problems created a shortage of the flu vaccines for the 2004–2005season. By comparison, statistical input in areas such as high-throughput screening,chemical development, formulation development, drug delivery, and assay development isbeing sought only when the scientists feel that statisticians could truly add value. Thismentality could limit statisticians’ contributions since researchers will not know how

Page 16: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 1 Statistics in Drug Development 3

statisticians could help unless they have previously worked with statisticians or have beenreferred to statisticians by their grateful colleagues. For example, scientists who are used toexperimenting with one factor at a time won’t know the value of factorial experiments.Similarly, even though statisticians well versed in Six Sigma and Design for Six Sigma arewell aware of the many applications of the Six Sigma principles, they need to actively sellthe applications to potential clients.

The non-clinical support model differs from that in the clinical area because of theusually large client-to-statistician ratio. As a result, after a statistician completes aparticular job, he/she often looks for opportunity to consolidate the techniques andinstitutionalize the tools for the client to use on a routine basis. The automation allowsstatisticians to focus on opportunities for new collaboration and developing newmethodologies for applications.

Non-clinical statisticians often work individually with their clients. Lendrem (2002)described them as “pioneers” because of the frequent needs to venture into unknown areasof new technology. Quantifying gene expression via the microarray technology is one suchexample. Another is industry’s (and government’s alike) investment in identifyingbiomarkers for testing mechanism of action of new molecular or biologic entities. In bothcases, the findings will have great clinical implications, but the work starts in the researchlaboratories and our non-clinical statisticians are the first to deal with the need to measure,to quantify, and to validate the measurements from the technical perspective.

Because of the small number of non-clinical statisticians in many pharmaceuticalcompanies, it is useful for non-clinical statisticians to form an inter-company network tobenefit mutual learning. Some of this networking has been in existence for some time. Inthe U.S., a CMC (Chemistry, Manufacturing, and Control) Statistical Expert Team wasformed in the late 60’s to focus on the chemistry and control issues related to themanufacturing of pharmaceutical products. Another example is the PharmacogenomicsStatistical Expert Team that was formed in the fall of 2003. Both teams are sanctioned byPhRMA and consist of statisticians from major pharmaceutical companies.

1.3 Statistical Support to Clinical TestingClinical testing is typically conducted in a staged fashion to explore the effect ofpharmaceutical products on humans. The investigation starts with pharmacokinetic andpharmacodynamic studies, followed by proof-of-concept and dose-ranging studies. Somespecialty studies such as drug effect on QT/QTc prolongation and drug-drug interactionsstudies are conducted at this early stage. The identification of common adverse reactionsand early signs of efficacy are the objectives of such trials. The early testings, ifsatisfactory, lead to the confirmatory phase where the efficacy and safety of the productcandidate are more thoroughly investigated in a more heterogeneous population.

Despite common statistical principles, different stages of clinical testing focus ondifferent statistical skill sets. For early proof-of-concept and dose-ranging efforts, studydesigns could be more flexible and the goal is to learn as efficiently and effectively aspossible. Adaptations, in terms of dose allocation, early termination, and study populationgive great flexibility to such trials. Extensive modeling that incorporates accumulatedlearning on a real-time basis can lead a sponsor to decision points in a more expeditedfashion. Because the purpose of this phase of development is primarily to generateinformation to aid internal decisions, the developers are freer to use innovative approachesas long as they can successfully defend the decisions that become the basis for laterdevelopment.

By comparison, statistical approaches for the confirmatory phase need to be carefullypre-planned, pre-specified, and followed in order to give credibility to the results. Apharmaceutical sponsor needs to decide a priori study designs, primary endpoints, primaryanalysis population, success criteria, handling of missing data, multiple comparisons, plus

Page 17: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

4 Pharmaceutical Statistics Using SAS: A Practical Guide

many others. ICH E9 (1998) gives a very detailed description of all aspects of trial designand analysis that a statistician should consider at this stage. When adaptation is planned,the rule needs to be clearly specified in advance. When interim analysis is anticipated, asponsor’s access to the interim results needs to be tightly controlled.

The confirmatory phase is the place where knowledge about a new molecular or biologicentity is solidified to support a target label. The knowledge, along with the approved label,becomes the basis for recommendations to prescribing physicians and the medicalcommunity. The confirmatory trials are also the place where the risk-benefit andcost-effectiveness of a new pharmaceutical product are first delineated. The greater numberof subjects studied at this stage gives a sponsor a decent chance to study adverse actionsthat have a rate between 0.1% and 1%. This phase overlaps somewhat with the life cyclemanagement phase where new indications are being explored and drug differentiation isbeing sought. If there is a post-marketing study commitment, additional studies will beinitiated to fulfill the conditions for approval.

Increasingly, statisticians are participating in promotion review and educationalcommunications to the general public. In addition, many statisticians contribute toactivities related to pharmacovigilance and epidemiology.

1.4 Battling a High Phase III Failure RateThe attrition rate of compounds in the pharmaceutical industry is extremely high. Settingaside compounds that fail the preclinical testing, it is generally recognized that less than12% of compounds entering into the human phase testing will eventually make it to themarket place. The rate is a composite figure formed as the product of the success rates ofpassing the Phase I testing, passing the Phase II testing, passing the Phase III testing, andpassing the regulatory review. Among failures at the various stages, Phase III attrition hasthe greatest impact. This is so not only because of all the accumulated resources expendedup to this point, but it is also because Phase III failure represents a great disappointmentto the sponsor, leaving the sponsor short of a defendable marketing application.

In a recent article, Chuang-Stein (2004) conducted a root cause analysis of the Phase IIIfailure rate that was reported to be running at the 50% level. This most recent figure ishigher than the 32% rate reported in DiMasi, Hansen, and Grabowski (2003).Chuang-Stein attributed the cause to three major factors: the candidate factor, the sponsorfactor, and the environmental factor. While we can’t dismiss the pipeline problem, and wehave admittedly very little control over the behaviors of some corporate decision-makers atthe highest level, many of the causes indeed relate to how clinical development is beingconducted and how decisions are made to move compounds through different phases of thedevelopment. Chuang-Stein discussed what statisticians could do to help reduce theattrition rate at the late stage. One area where the methodology is well developed andstatisticians could make immediate contributions is the judicious use of adaptive designs,or at least group sequential designs, in Phase III trials. The goal of such designs is to givethe Phase III trials the best chance for success or to terminate them early if the trials arenot likely to meet their objectives. Implicit in such designs is the inclusion of more decisionpoints based on interim results to allow evidence-based decisions. The need to incorporateregular decision points is not limited to Phase III testing. It should be part of every stageof the drug development continuum. These decision points serve as reality checks on thelong and costly development journey.

The industry is at a crossroad, and changes are critically needed. Statisticians shouldtake advantage of the challenges and fully engage themselves in looking for better ways tosupport clinical development of pharmaceutical products.

Page 18: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 1 Statistics in Drug Development 5

1.5 Do Statisticians Count?In a soul-searching article, Andy Grieve (2002) asks whether statisticians count. Eventhough the number of statisticians working in the pharmaceutical industry has increasedby 50-fold since the late 70’s, Grieve felt that the influence statisticians had in theirrespective companies had not increased proportionally. Grieve looked at the barriers thatprevented statisticians from contributing as much as they could and offered some solutions.

Particularly noteworthy is the assertion that it is the statistician, and not statistics,that is important. Statistics, as a discipline, does not influence, does not persuade, does notdesign studies, does not analyze data, does not interpret findings, and does not reportresults. Statisticians are the ones who make the discipline meaningful by doing all of theabove. In other words, statisticians, through their own behavior and communication,spread the statistical principles and promote the statistical thinking. So, when we discussthe successful use of statistics in drug development, we need to bear in mind that asstatisticians working in the pharmaceutical industry, we need to be the champions for suchcauses through our passion for the statistics profession.

1.6 Emerging OpportunitiesThe Critical Path initiative document describes many opportunities to improve theefficiency of product development. We will mention just a few here. On better tools toassess a compound’s safety, FDA states the need for new techniques to evaluate drug livertoxicity, new methods to identify gene therapy risk, better predictors of human immuneresponses to foreign antigens, new methods to further enhance the safety of transplantedhuman tissues, and efficient protocols for qualifying biomaterials. On better tools todemonstrate the medical utility of a compound, FDA shares the agency’s successfulexperience with biomarkers in HIV infection and duodenal ulcer healing. FDA states theneed for more biomarkers and surrogate markers that can guide product development. Inaddition, FDA discusses the need for better animal models to combat bioterrorism, moreclinically relevant endpoints, better imaging technologies, more innovative designs andanalysis methods, and the need for implementing the concept of model-based drugdevelopment. The latter involves building mathematical and statistical characterization ofthe time course of the disease and the drug effect, using available clinical data.

The Critical Path initiative document also discusses the need for better methods tocharacterize, standardize, control, and manufacture medical products on the commercialscale. Since manufacturing expenses could exceed the research and developmentinvestment, there is a need for a better validation process that follows the risk-basedinspection paradigm advocated by the FDA in recent months. The latter includes moreattention to setting specifications and shifting from detailed data analysis to overall processquality assessment. The same philosophy suggests moving toward acceptance of aprobabilistic definition, rather than a pass or fail on the manufacturing process. Mostimportant, FDA wants to encourage the manufacturers to integrate state-of-the-art scienceand technology into their manufacturing processes.

Following the issuance of the Critical Path initiative document, different centers withinthe FDA have further identified areas for innovations and have presented opportunities tothe FDA’s Science Board on November 5, 2004. Since May 2004, many workshops havedirected at least part of their agenda towards more efficient and effective ways to test anddevelop new treatments. Common to many of the discussions are the needs to applyquantitative thinking and techniques. Taking the clinical phase of product development asan example, we see that a major emphasis is to use mathematical and statistical models tohelp guide drug development and approval. The central idea is to pool data from multipletrials to augment our knowledge base and actively incorporate such knowledge insubsequent studies. Interestingly enough, the concept of pooling data has now been

Page 19: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

6 Pharmaceutical Statistics Using SAS: A Practical Guide

extended to pooling data of drugs that belong to the same class. The pooling ofinformation across companies, while challenging, will undoubtedly facilitate the collectivelearning of the pharmaceutical industry.

The opportunities for statisticians to make substantial contributions at the strategiclevel are beyond what one could have imagined 20 years ago. Along with the opportunitiescome expectations that statisticians will help solve the puzzle faced by modern-dayscientists in the pharmaceutical industry.

1.7 SummaryStatistics, as a discipline, has broadened its scope significantly over the past 20 years.Wherever there is a need for quantification, statistics has a role. The ability to think interms of variability, to separate signals from noise, to control sources of bias and variation,and to optimize under given conditions, makes statisticians a valuable partner in thedevelopment of new pharmaceutical and biological products.

Mining historical data to add to our cumulative knowledge is a low-cost and high-yieldactivity. Many companies realize the value of this activity and are actively pursuing it. Forexample, Bristol-Myers Squibb (Pink Sheet, December 13, 2004) formed a discoverytoxicology group and retrospectively analyzed approximately 100 development compoundsthat failed during a 12-year period. Bristol-Myers Squibb hoped to use the acquiredknowledge to decide what assays and technology to implement early to reduce compoundattrition. Bristol-Myers Squibb concluded that a combination of in vitro, in vivo, and insilico techniques was needed to improve productivity and reduce attrition. According to thesame report in the Pink Sheet (December 13, 2004), other pharmaceutical companies havereached similar conclusions.

Data mining is also expected to help us look for better predictors for hepatotoxicity andcardiovascular toxicity such as Torsade de pointes. Data mining examples can go on andon. We can’t think of any scientists who are more poised and qualified to lead thisdata-based learning endeavor than statisticians!

The challenge is definitely on us, statisticians!

ReferencesChuang-Stein, C. (2004). “Seize the opportunities.” Pharmaceutical Statistics. 3, 157–159.DiMasi, J., Hansen, R.W., Grabowski, H.G. (2003). “The price of innovation: new estimates of drug

development costs.” Journal of Health Economics. 22, 151–185.“FDA ‘Critical Path’ May Lead To Changes In Dosing, Active-Control Trials.” December 13, 2004.

Pink Sheet. 31.Grieve, A. (2002). “Do statisticians count? A personal view.” Pharmaceutical Statistics. 1, 35–43.International Conference of Harmonization of Pharmaceuticals for Human Use (ICH) E9. (1998).

“Statistical Principles for Clinical Trials.” Available at http://www.ich.org.Lendrem, D. (2002). “Statistical support to non-clinical.” Pharmaceutical Statistics. 1, 71–73.Pharmaceutical Research and Manufacturers of America (PhRMA) Report. (2006). Value of

Medicines. Available at http://www.phrma.org/education/.The Critical Path to New Medical Products. (March 2004). Available at

http://www.fda.gov/oc/initiatives/criticalpath/whitepaper.html.

Page 20: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2

Modern ClassificationMethods for Drug Discovery

Kjell JohnsonWilliam Rayens

2.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .7

2.2 Motivating Example . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

2.3 Boosting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10

2.4 Model Building . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .27

2.5 Partial Least Squares for Discrimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33

2.6 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42

This chapter focuses on two modern methods for classification: boosting and partial leastsquares for discrimination. For each of these methods, we provide detailed backgroundinformation about their theory as well as instruction and guidance on their implementationin SAS. Finally, we apply these methods to a common drug discovery-type data set andexplore the performance of the two methods.

2.1 IntroductionDrug discovery teams are often faced with data for which the samples have beencategorized into two or more groups. For example, early in the drug discovery process, highthroughput screening is used to identify compounds’ activity status against a specificbiological target. At a subsequent stage of discovery, screens are used to measurecompounds’ solubility, permeability, and toxicity status. In other areas of drug discovery,information from animal models on disease classification, survival status, and occurrence ofadverse events is obtained and scrutinized. Based on these categorizations, teams mustdecide which compounds to pursue for further development.

In addition to the categorical response, discovery data often contain variables thatdescribe features of the samples. For example, many computational chemistry softwarepackages have the ability to generate structural and physical-property descriptors for anydefined set of compounds. In genomics and proteomics, expression profiles can be measuredon tissue samples.

Kjell Johnson is Associate Director, Nonclinical Statistics, Pfizer, USA. William Rayens is Professor, Department of Statis-tics, University of Kentucky, USA.

Page 21: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

8 Pharmaceutical Statistics Using SAS: A Practical Guide

Given data that contain descriptors and a categorical response, teams desire to uncoverrelationships between the descriptors and response that can provide them with scientificintuition and help them predict the classification of future compounds or samples.

In drug discovery, data sets are often large and over-described (more descriptors existthan samples or highly correlated descriptors). And often, the relationships betweendescriptors and classification grouping are complex. That is, the different classes of samplescannot be easily separated by a line or hyperplane. Hence, methods that rely on invertingthe covariance matrix of the descriptors, or methods that find the best separatinghyperplane are not effective for this type of data. Traditional statistical methods such aslinear discriminant analysis or logistic regression are both linear classification methods andrely on the covariance matrix of the descriptors. For the reasons mentioned previously,neither is optimal for modeling many discovery type data sets.

In this situation, one approach is to reduce the descriptor space in a way that stabilizesthe covariance matrix. Principal component analysis, variable selection techniques such asgenetic algorithms, or ridging approaches are popular ways to obtain a stable covariancematrix. When the matrix is stable, traditional discrimination techniques can beimplemented. In each of these approaches, the dimension reduction is performedindependently of the discrimination. An alternative approach, partial least squares forlinear discrimination, has the added benefit of simultaneously reducing dimension whilefinding the optimal classification rule.

Another promising classification method is support vector machines (Vapnik, 1996). Inshort, support vector machines seek to find a partition through the data that maximizesthe margin (the space between observations from separate classes), while minimizingclassification error. For data in which classes are clearly separable into different groups, itis possible to maximize the margin while minimizing classification error, regardless of thecomplexity of the boundary. However, for data whose classes overlap, maximizing themargin and minimizing classification error are competing constraints. In this case,maximizing the margin produces a smoother classification boundary, while minimizingclassification error produces a more flexible boundary. Although support vector machinesare an effective classification tool, they can easily overfit a complex data set and can bedifficult to interpret.

Recursive partitioning seeks to find individual variables from the original data thatpartition the samples into more pure subsets of the original data (Breiman et al., 1984).Thus, a recursive partition model is a sequence of decision rules on the original variables.Because recursive partitioning selects only one variable at each partition, it can be used tomodel an overdetermined data set. This method can also find a more complex classificationboundary because it partitions the data into smaller and smaller hypercubes. While arecursive partition model is easy to interpret, its greedy nature can prevent it from findingthe optimal classification model for the data.

More recently, methods which combine classifiers, known as ensemble techniques, havebeen shown to outperform many individual classification techniques. Popular ensemblemethods include bagging (Breiman, 1996), ARCing (Breiman, 1998), and boosting (Freundand Schapire, 1996a), which have been shown to be particularly effective classification toolswhen used in conjunction with recursive partitioning.

Because of their popularity, effectiveness, and ease of implementation in SAS, we willlimit the focus of this chapter to boosting and partial least squares for discrimination.

In Section 2.2, we will motivate our methods with an example of a typical drugdiscovery data set. Section 2.3 will explore boosting and its implementation in SAS as wellas its effectiveness on the motivating example and Section 2.4 will review model buildingtechniques. Section 2.5 will discuss the use of partial least squares for discrimination andSAS implementation issues, and apply this method to the motivating example.

Page 22: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 9

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

2.2 Motivating ExampleGenerally speaking, permeability is the ability of a molecule to cross a membrane. In thebody, key membranes exist in the intestine and brain, and are composed of layers ofmolecules and proteins organized in a way to prevent harmful substances from crossingwhile allowing essential substances to pass through. The intestine, for instance, allowssubstances such as nutrients to pass from the gut into the blood stream. Anothermembrane, the blood-brain barrier, prevents detrimental substances from crossing theblood stream into the central nervous system. While a molecule may have the correctcharacteristics to be effective against a specific disease or condition, it may not have thecorrect characteristics to pass from the gut into the blood stream or from the blood streaminto the central nervous system. Therefore, if a potentially effective compound is notpermeable, then its effectiveness may be compromised.

Because a compound’s permeability status is critically important to its success,pharmaceutical companies would like to identify poorly permeable compounds as early aspossible in the discovery process. These compounds can then be eliminated from follow-up,or can be modified in an attempt to improve permeability while keeping their potentialtarget effectiveness.

To measure a compound’s ability to permeate a biological membrane, several in-vitroassays such as PAMPA and Caco-2 have been developed (Kansy et al., 1998). In each ofthese assays, cell layers are used to simulate a body-like membrane. A compound is thenplaced into solution and is added to one side of the membrane. After a period of time, thecompound concentration is measured on the other side of the membrane. Compounds withpoor permeability will have low compound concentration, while compounds with highpermeability will have high compound concentration.

These screens are often effective at identifying the permeability status of compounds,but the screening process is moderately labor- and material-intensive. At a typicalpharmaceutical company, resources are limited to screening only a few hundred compoundsper week. To process more compounds would require more resources. Alternatively, wecould attempt to build a model that would predict permeability status.

As mentioned above, biological membranes are a complex layer of molecules andproteins. To pass through a membrane, a substance must have an appropriate chemicalcomposition. Therefore, to build a predictive model of permeability status we shouldinclude appropriate chemical measurements for each compound.

For the example in this chapter, we have collected data on 354 compounds (the PERMYdata set can be found on the book’s companion Web site). For each compound we have itsmeasured permeability status (y = 0 is not permeable and y = 1 is permeable), and we haveused in-house software to compute 71 molecular properties that are theoretically related topermeability (i.e., hydrogen bonding, polarity, molecular weight, etc.). For proprietaryreasons, these descriptors have been blinded and are labeled as X1, X2, . . . , X71.

As is common with many drug discovery data sets, the descriptor matrix for the data isoverdetermined (that is, at least one variable is linearly dependent on one or more of theother variables). To check this in SAS, we can use the PRINCOMP procedure to generatethe eigenvalues of the covariance matrix. Recall that a full rank covariance matrix will haveno zero eigenvalues. Program 2.1 generates the eigenvalues of the covariance matrix.

Page 23: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

10 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 2.1 Permeability data set: Computation of the eigenvalues of the covariance matrix

proc princomp data=permy cov n=71 outstat=outpca noprint;var x1-x71;

data outpca;set outpca(where=(_type_="EIGENVAL"));keep x1-x71;

proc transpose data=outpca out=outpca_t(drop=_name_) prefix=eig;var x1-x71;

proc print data=outpca_t;run;

Output from Program 2.1

Obs eig1

1 531907.982 202322.503 39308.424 22823.335 9187.106 5087.367 2660.498 1229.599 814.6210 495.17

62 .00000306363 .00000085764 .00000020965 .00000014966 067 068 069 070 071 0

Output 2.1 lists the first 10 and last 10 eigenvalues of the covariance matrix computedfrom the permeability data set. Notice that the covariance matrix for the permeability datais not full rank. In fact, six eigenvalues are zero, while more than 20 are less than 0.01.This implies that the covariance matrix is not invertible and methods that rely on theinverse of the covariance matrix will have a difficult time with these data.

Another common situation in which the descriptor matrix can be overdetermined occurswhen we have more descriptors than observations. Methods that rely on a full-rankcovariance matrix of the descriptors, such as linear discriminant analysis, will fail with thistype of data. Instead, we must either remove the redundancy in the data or employmethods, such as boosting or partial least squares for linear discrimination, that can lookfor predictive relationships in the presence of overdetermined data.

2.3 BoostingSeveral authors have written thorough summaries of the historical development of boosting(see, e.g., Friedman et al., 2000; Schapire, 2002) For completeness of this work, we providea brief overview of boosting and include recent findings.

Page 24: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 11

2.3.1 Evolution of BoostingThe concept of boosting originated in the machine learning community where Kearns andValiant (1989) explored weak learning algorithms (algorithms that can classify objectsbetter than random) and strong learning algorithms (algorithms that can classifyaccurately). The question of the relationship between weak and strong learning algorithmswas initially posed, and was addressed soon after by Schapire (1990). In his work, Schapireshowed that if a concept was weakly learnable it was also strongly learnable, and heformulated the first algorithm to “boost” a weak learner into a strong learner. Freund(1995) improved upon Schapire’s initial algorithm by reducing its complexity andincreasing its efficiency. However, both Schapire’s algorithm and Freund’s algorithms weredifficult to implement in practice.

To overcome these practical implementation problems, Freund and Schapire (1996a)collaborated to produce the well-known and widely applied AdaBoost algorithm describedbelow (the notation used in this algorithm is defined in Table 2.1).

1. Let w1,1 = · · · = w1,n = 1/n.2. For t = 1, 2, . . . , T do:

(a) Fit ft using the weights, wt,1, . . . , wt,n, and compute the error, et.(b) Compute ct = ln((1 − et)/et).(c) Update the observation weights:

wt+1,i = wt,i exp(ctIt,i)/n∑

j=1

(wt,j exp(ctIt,j)), i = 1, . . . , n.

3. Output the final classifier:

yi = F (xi) = sign

(T∑

t=1

ctft(xi)

).

Table 2.1 Notation Used in the AdaBoost Algorithm

i Observation number, i = 1, 2, . . . , n.t Stage number, t = 1, 2, . . . , T .xi A p-dimensional vector containing the quantitative variables of the ith

observation.yi A scalar quantity representing the class membership of the ith observation,

yi = −1 or 1.ft The weak classifier at the tth stage.ft(xi) The class estimate of the ith observation using the tth stage classifierwt,i The weight for the ith observation at the tth stage,

∑i wt,i = 1.

It,i Indicator function, I(ft(xi) �= yi).et The classification error at the tth stage,

∑i wt,iIt,i.

ct The weight of ft.sign(x) = 1 if x ≥ 0 and = −1 otherwise.

In short, AdaBoost generates a sequentially weighted additive agglomeration of weakclassifiers. In each step of the sequence, AdaBoost attempts to find the best classifieraccording to the current distribution of observation weights. If an observation is incorrectlyclassified with the current distribution of weights, the observation receives more weight inthe next sequence, while correctly classified observations receive less weight in the nextiteration. In the final agglomeration, classifiers which are accurate predictors of the original

Page 25: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

12 Pharmaceutical Statistics Using SAS: A Practical Guide

data set receive more weight, whereas weak classifiers that are poor predictors receive lessweight. Thus, AdaBoost uses a sequence of simple weighted classifiers, each forced to learna different aspect of the data, to generate a final, comprehensive classifier, which oftenclassifies better than any individual classifier.

Friedman, Hastie, and Tibshirani (2000) dissected the AdaBoost algorithm and revealedits statistical connections to loss functions, additive modeling, and logistic regression.Specifically, AdaBoost can be thought of as a forward stagewise additive model that seeksto minimize an exponential loss function:

e−yF (x),

where F (x) denotes the boosted classifier (i.e., the classifier defined in Step 3 of thealgorithm). Using this framework, Friedman, Hastie, and Tibshirani (2000) generalized theAdaBoost algorithm to produce a real-valued prediction (Real AdaBoost) andcorresponding numerically stable version (Gentle AdaBoost). In addition, they replaced theexponential loss function with a function more commonly used in the statistical field forbinary data, the binomial log-likelihood loss function, and named this method LogitBoost.Table 2.2 provides a comparison of these methods.

Generalized Boosting Algorithm of Friedman, Hastie, and Tibshirani (2000)1. Initialize the observation weights, w1,i, and response for model, ywork

i .2. For t = 1, 2, . . . , T do:

(a) Fit the model yworki = f(xi) using the weights, wt,i.

(b) Compute ct(xi), a contribution of xi at stage t, to the final classifier.(c) Update the weights, wt,i. Update ywork

i (for LogitBoost only).3. Output the final classifier:

yi = F (xi) = sign

(T∑

t=1

ct(xi)

).

2.3.2 Weak Learning Algorithms and Effectiveness of BoostingAs mentioned in the previous section, boosting can be applied to any classification methodthat classifies better than random. By far, the most popular and effective weak learners aretree-based methods, also known as recursive partitioning. Several authors have providedexplanations of why recursive partitioning can be boosted to produce effective classifiers(see, for example, Breiman, 1998). In short, boosting is effective for classifiers that areunstable (produce different models when given different training data from the samedistribution), but when combined, get approximately the correct solution (i.e., have lowbias). This breakdown is commonly known as the bias-variance trade-off (see, for example,Bauer and Kohavi, 1999). The importance of using an unstable classifier can be easily seenby examining the AdaBoost algorithms further. Specifically, if a classifier misclassifies thesame observations in consecutive iterations, then AdaBoost will be prevented from findingadditional structure in the data.

To explain further, let wt,i represent the weight of the ith observation at the tth stage ofAdaBoost. Without loss of generality, suppose that the first m observations aremisclassified and

∑ni=1 wt,i = 1. By the AdaBoost algorithm, the updated weights are

wt+1,i = wt,iectIt,i

⎛⎝ n∑j=1

wt,jectIt,j

⎞⎠−1

.

Page 26: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 13

Table 2.2 Comparison of the Generalized Boosting Algorithms

AlgorithmAlgorithmparameter AdaBoost Real AdaBoost

w1,i 1/n 1/n

yi ∈ {−1, 1} ∈ {−1, 1}ywork

i yi yi

ft Any weak classification method Any weak classification methodthat predicts a binary that predicts class probability

response: ft(xi) ∈ {−1, 1} P (yi = 1|xi), ft(xi) ∈ [0, 1]

ct(xi) αtft(xi), where αt = ln1 − et

et,

12

lnft(xi)

1 − ft(xi)et =

∑ni=1 wt,iI(ft(xi) �= yi)

wt+1,iwt,i exp(αtI(ft(xi) �= yi))∑ni=1 wt,i exp(αtI(ft(xi) �= yi))

wt,i exp(−yict(xi))∑ni=1 wt,i exp(−yict(xi))

Gentle AdaBoost LogitBoostw1,i 1/n 1/4yi ∈ {−1, 1} ∈ {0, 1}

yworki yi 4yi − 2ft Any method that Any method that

predicts a continuous predicts a continuousresponse: ft(xi) ∈ R response: ft(xi) ∈ R

ct(xi) ft(xi) ft(xi)

wt+1,iwt,i exp(−yift(xi))∑ni=1 wt,i exp(−yift(xi))

[exp(Ft(xi)) + exp(−Ft(xi))]−2,

where Ft(xi) = (1/2)∑t

j=1 fj(xi),

yworki =

(yi − 1

1 + exp(−2Ft(xi))

)/wt+1,i

Next, suppose the first m observations are again misclassified in the (t + 1)st stage. Then,

et+1 =m∑

j=1

wt,ject

⎛⎝ m∑j=1

wt,ject +

n∑j=m+1

wt,j

⎞⎠−1

.

Notice that

m∑j=1

wt,ject =

1 − et

et

m∑j=1

wt,j =m∑

j=1

wt,j

⎛⎝1 −m∑

j=1

wt,j

⎞⎠⎛⎝ m∑j=1

wt,j

⎞⎠−1

=n∑

j=m+1

wt,j .

Therefore, et+1 = 0.5 and ct+1 = 0. This implies that the weights for iteration t + 2 will bethe same as the weights from iteration t + 1, and the algorithm will choose the same model.This fact will prevent the algorithm from learning any more about the relationship betweenthe descriptor space and response classification. Hence, methods that are stable, such aslinear discriminant analysis, k-nearest neighbors, and recursive partitions with manyterminal nodes, will not be greatly improved by boosting. But, methods such as neuralnetworks (Freund and Schapire, 1996b), Naive-Bayes (Bauer and Kohavi, 1999), and

Page 27: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

14 Pharmaceutical Statistics Using SAS: A Practical Guide

recursive partitions with few terminal nodes (also referred to as stumps) will be improvedby boosting. For example, Optiz and Maclin (1999) compared boosting neural networks toboosting decision trees on twenty-three empirical data sets. For a majority of the data sets,boosting neural networks or decision trees produced a more accurate classifier than anyindividual neural network or decision tree, respectively. However, Optiz and Maclinillustrated that the performance of boosting was data dependent; for several data sets aboosted neural network performed worse than an individual neural network. Additionally,Bauer and Kohavi (1999) illustrated that boosting a Naive-Bayes classifier improvesclassification, but can increase the variance of the prediction.

Because of the success of boosting with recursive partitioning using few terminal nodes,in the next section we will explore the implementation of boosting using stumps, arecursive partition with one split.

2.3.3 Implementation of Boosting in SASThere are at least two ways to implement boosting in SAS. Conveniently, boosting can bedirectly implemented in SAS Enterprise Miner software. However, the use of SASEnterprise Miner requires the purchase of this module. Boosting can also be implementedthrough more commonly and widely available SAS software such as Base SAS, SAS/STAT,and SAS/IML. This section will focus on the development of boosting code using widelyavailable SAS software. But, we begin this section by exploring a few details of boosting inSAS Enterprise Miner.

2.3.4 Implementation with SAS Enterprise MinerIn SAS Enterprise Miner, an ensemble classifier is quite easy to create, and can begenerated with a minimum of four nodes (see Table 2.3). SAS documentation providesthorough step-by-step directions for creating various ensembles of classifiers in theEnsemble Node Help section; we refer you to this section for details on how to implementan ensemble model. Here, we highlight a few key facts about boosting in SAS EnterpriseMiner.

Table 2.3 Required Nodes to Perform Boosting in SAS Enterprise Miner

Node Options

1. Input Data Source Select target (response) variable2. Group Processing Mode = Weighted resampling for boosting3. Model Select Tree model for a boosted recursive partition4. Ensemble Ensemble node = Boosting

To generate an ensemble model, the software requires the Input Data Source, GroupProcessing, Model, and Ensemble nodes. In the General tab under Group Processing, onemust select Weighted resampling for boosting and must specify the number of loops(iterations) to perform. Then, in the ensemble node, boosting must be chosen as thesetting. It is important to note that SAS Enterprise Miner performs boosting by buildingtrees on weighted resamples of the observations rather than buildings trees based onweighted observations. This type of boosting is more commonly referred to as adaptiveresampling and combining (ARCing) and was developed by Breiman (1998). In fact, theSAS Enterprise Miner implementation of boosting is only a slight modification of Breiman’sArc-x4 algorithm. While boosting and ARCing have the same general flavor, Friedman,Hastie and Tibshirani (2000) indicate that boosting via weighted trees generally performsbetter than ARCing. Hence, a distinction should be made between the two methods.

Unfortunately, the constructs within SAS Enterprise Miner make it extremely difficultto implement boosting with weighted trees and any of the boosting algorithms from

Page 28: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 15

Table 2.2. Instead, we implement the generalized boosting algorithms directly throughmore commonly used SAS software.

2.3.5 Implementation of Boosting in Base SASFor our implementation of boosting, we will use stumps—recursive partitions with onesplit—as the weak learner. We begin this section by focusing on the construction of code tofind an optimal recursive partition in a set of data.

EXAMPLE: Recursive PartitioningRecursive partitioning seeks to find successive partitions of the descriptor space thatseparate the observations into regions (or nodes) of increasingly higher purity of theresponse. As an illustration, consider a data set that contains 100 four-dimensional points,(x1, x2, y, w), where y is the class identifier and w is the weight. The data set is generatedby Program 2.2. A plot of the generated data points is displayed in Figure 2.1.

Program 2.2 Simulated data in the recursive partitioning example

data example1;do i=1 to 100;

x1 = 10*ranuni(1);x2 = 10*ranuni(2);if (x1<5 and x2<1.5) then y=0;if (x1<5 and x2>=1.5) then y=1;if (x1>=5 and x2<7.5) then y=0;if (x1>=5 and x2>=7.5) then y=1;w=1;output;

end;* Vertical axis;axis1 minor=none label=(angle=90 "X2") order=(0 to 10 by 1) width=1;* Horizontal axis;axis2 minor=none label=("X1") order=(0 to 10 by 1) width=1;symbol1 i=none value=circle color=black height=4;symbol2 i=none value=dot color=black height=4;proc gplot data=example1;

plot x2*x1=y/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

For this example, we desire to find a single partition of the data that creates two newregions of highest possible purity. Of course, the best partition is dependent on the measureof region impurity. An intuitive measure of impurity is misclassification error, but a moreaccepted impurity measure is the Gini Index. Let pj represent the proportion ofobservations of Class j in a node. For a two-class problem, the misclassification error of anode is defined as min(p1, p2), and the Gini Index is defined as 2p1p2. The total measure ofimpurity is a weighted sum of the impurity from each node, where each node is weightedby the proportion of observations in that node, relative to the total number of observationsfrom its parent node.

For the EXAMPLE1 data set, a few possible partitions clearly stand out (seeFigure 2.1):

x1 = 5, x2 = 7.5 or x2 = 1.5.

Classification using these cut-points yields the results in Table 2.4. The correspondingmeasures of misclassification error and Gini Index can be found in Table 2.5.

Page 29: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

16 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 2.1 Plot of the data in the recursive partitioning example

X2

0

1

2

3

4

5

6

7

8

9

10

X1

0 1 2 3 4 5 6 7 8 9 10

Table 2.4 Classification Results

Class x1 ≥ 5 x1 < 5 x2 < 7.5 x2 ≥ 7.5 x2 < 1.5 x2 ≥ 1.5

y = 0 40 11 51 0 14 37y = 1 7 42 32 17 0 49

Table 2.5 Computation of the Misclassification Error and Gini Index

Partition Misclassification error Gini index

x1 ≥ 5 min(7/47, 40/47) = 0.15 2(7/47)(40/47) = 0.25x1 < 5 min(11/53, 42/53) = 0.21 2(11/53)(42/53) = 0.33Total (0.47)(0.15) + (0.53)(0.21) = 0.18 (0.47)(0.25) + (0.53)(0.33) = 0.29

x2 ≥ 7.5 min(32/83, 51/83) = 0.39 2(32/83)(51/83) = 0.47x2 < 7.5 min(0/17, 17/17) = 0 2(0/17)(17/17) = 0Total (0.83)(0.39) + (0.17)(0) = 0.32 (0.83)(0.47) + (0.17)(0) = 0.39

x2 ≥ 1.5 min(37/86, 49/86) = 0.43 2(37/86)(49/86) = 0.49x2 < 1.5 min(0/14, 14/14) = 0 2(0/14)(14/14) = 0Total (0.14)(0) + (0.86)(0.43) = 0.37 (0.14)(0) + (0.86)(0.49) = 0.42

Table 2.5 shows that the partition at x1 = 5 yields the minimum value for both totalmisclassification error (0.18) and the Gini Index (0.29).

Unequally Weighted ObservationsAs defined above, misclassification error and Gini Index assume that each observation isequally weighted in determining the partition. These measures can also be computed for

Page 30: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 17

unequally weighted data. Define

p∗j =

n∑i=1

wiI(yi = j)

(n∑

i=1

wi

)−1

.

Then, for the two-class problem, the misclassification error and Gini Index are min(p∗1, p

∗2)

and 2p∗1p

∗2, respectively.

While three partitions for the EXAMPLE1 data set visually stand out in Figure 2.1,there are 198 possible partitions of these data. In general, the number of possible partitionsfor any data set is the number of dimensions multiplied by one fewer than the number ofobservations. And, to find the optimal partition one must search through all possiblepartitions. Using data steps and procedures, we can construct basic SAS code toexhaustively search for the best partition using the impurity measure of our choice, andallowing for unequally weighted data.

%Split MacroThe %Split macro searches for the optimal partition using the Gini Index (the macro isgiven on the book’s companion Web site). The input parameters for %Split are INPUTDSand P. The INPUTDS parameter references the data set for which the optimal split will befound, and this data set must have independent descriptors named X1, X2, . . . , XP, avariable named W that provides each observation weight, and a response variable named Ythat takes values 0 and 1 for each class. The user must also specify the P parameter, thenumber of independent descriptors. Upon being called, %Split produces an output data setnamed OUTSPLIT that contains the name of the variable and split point that minimizesthe Gini Index.

Program 2.3 calls the %Split macro to find the optimal binary split in the EXAMPLE1data set using the Gini Index.

Program 2.3 Optimal binary split in the recursive partitioning example

%split(inputds=example1,p=2);proc print data=outsplit noobs label;

format gini cutoff 6.3;run;

Output from Program 2.3

Gini Best Bestindex variable cutoff

0.293 x1 5.013

Output 2.3 output identifies x1 = 5.013 as the split that minimizes the Gini Index. TheGini Index associated with the optimal binary split is 0.293.

While the %Split macro is effective for finding partitions of small data sets, its loopingscheme is inefficient. Instead of employing data steps and procedures to search for theoptimal split, we have implemented a search module named IML SPLIT that is based onSAS/IML and that replaces the observation loop with a small series of efficient matrixoperations.

AdaBoost Using the IML SPLIT Module

The AdaBoost algorithm described in Section 2.3.1 is relatively easy to implement usingthe IML SPLIT module as the weak learner (see the %AdaBoost macro on the book’s

Page 31: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

18 Pharmaceutical Statistics Using SAS: A Practical Guide

companion Web site). Like the %Split macro, %AdaBoost has input parameters ofINPUTDS and P, as well as ITER, which specifies the number of boosting iterations toperform. %AdaBoost creates a data set called BOOST that contains information abouteach boosting iteration. The output variables included in the BOOST data set are definedbelow.

• ITER is the boosting iteration number.• GINI VAR is the descriptor number at the current iteration that minimizes the Gini

Index.• GINI CUT is the cut-point that minimizes the Gini Index.• P0 LH is the probability of class 0 for observations less than GINI CUT.• P1 LH is the probability of class 1 for observations less than GINI CUT.• C L is the class label for observations less than GINI CUT.• P0 RH is the probability of class 0 for observations greater than GINI CUT.• P1 RH is the probability of class 1 for observations greater than GINI CUT.• C R is the class label for observations greater than GINI CUT.• ALPHA is the weight of the cut-point rule at the current boosting iteration.• ERROR is the misclassification error of the cumulative boosting model at the current

iteration.• KAPPA is the kappa statistic of the cumulative boosting model at the current iteration.

To illustrate the %AdaBoost macro, again consider the EXAMPLE1 data set.Program 2.4 calls %AdaBoost to classify this data set using ten iterations. The programalso produces plots of the misclassification error and kappa statistics for each iteration.

Program 2.4 AdaBoost algorithm in the recursive partitioning example

%AdaBoost(inputds=example1,p=2,iter=10);proc print data=boost noobs label;

format gini_cut p0_lh p1_lh p0_rh p1_rh c_r alpha error kappa 6.3;run;

* Misclassification error;* Vertical axis;axis1 minor=none label=(angle=90 "Error") order=(0 to 0.3 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 10 by 1) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=boost;

plot error*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

* Kappa statistic;* Vertical axis;axis1 minor=none label=(angle=90 "Kappa") order=(0.5 to 1 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 10 by 1) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=boost;

plot kappa*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

Page 32: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 19

Output from Program 2.4

iter gini_var gini_cut p0_LH p1_LH c_L p0_RH p1_RH c_R alpha error kappa

1 1 5.013 0.208 0.792 1 0.851 0.149 0.000 1.516 0.180 0.6402 2 6.137 0.825 0.175 0 0.082 0.918 1.000 1.813 0.230 0.5373 2 1.749 1.000 0.000 0 0.267 0.733 1.000 1.295 0.050 0.9004 1 5.013 0.272 0.728 1 0.877 0.123 0.000 1.481 0.050 0.9005 2 7.395 0.784 0.216 0 0.000 1.000 1.000 1.579 0.000 1.0006 2 1.749 1.000 0.000 0 0.228 0.772 1.000 1.482 0.050 0.9007 1 5.013 0.266 0.734 1 0.875 0.125 0.000 1.483 0.000 1.0008 2 7.395 0.765 0.235 0 0.000 1.000 1.000 1.462 0.000 1.0009 2 1.749 1.000 0.000 0 0.233 0.767 1.000 1.457 0.000 1.000

10 1 5.013 0.273 0.727 1 0.871 0.129 0.000 1.451 0.000 1.000

Output 2.4 lists the BOOST data set generated by the %AdaBoost macro. For the firstiteration, x1 minimizes the Gini index at 5.013. For observations with x1 < 5.013, theprobability of observing an observation in Class 0 is 0.208, while the probability ofobserving an observation in Class 1 is 0.792. Hence the class label for observations takingvalues of x1 < 5.013 is 1. Similarly, for observations with x1 > 5.013, the probabilities ofobserving an observation in Class 0 and Class 1 are 0.851 and 0.149, respectively.Therefore, the class label for observations taking values of x1 > 5.013 is 0. The stage weightfor the first boosting iteration is represented by ALPHA= 1.516, while the observedclassification error and kappa statistics are 0.18 and 0.64, respectively. Notice that theAdaBoost algorithm learns the structure of the relationship between the descriptor spaceand classification vector in seven iterations (the misclassification error and kappa statisticdisplayed in Figure 2.2 reach a plateau by the seventh iteration), and in these iterations thealgorithm quickly identifies cut-points near the constructed splits of x1 = 5, x2 = 7.5, andx2 = 1.5.

Figure 2.2 Misclassification error (left panel) and kappa statistic (right panel) plots for the EXAMPLE1 dataset using the %AdaBoost macro

Err

or

0.0

0.1

0.2

0.3

Iteration

1 2 3 4 5 6 7 8 9 10

Kap

pa

0.5

0.6

0.7

0.8

0.9

1.0

Iteration

1 2 3 4 5 6 7 8 9 10

Program 2.5 models the permeability data of Section 2.2 with AdaBoost and plots themisclassification error and kappa statistics at each iteration.

Page 33: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

20 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 2.5 AdaBoost algorithm in the permeability data example

%AdaBoost(inputds=permy,p=71,iter=50);proc print data=boost noobs label;

format gini_cut p0_lh p1_lh p0_rh p1_rh c_r alpha error kappa 6.3;run;

* Misclassification error;* Vertical axis;axis1 minor=none label=(angle=90 "Error") order=(0 to 0.3 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 51 by 10) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=boost;

plot error*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

* Kappa statistic;* Vertical axis;axis1 minor=none label=(angle=90 "Kappa") order=(0.5 to 1 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 51 by 10) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=boost;

plot kappa*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

Output from Program 2.5

iter gini_var gini_cut p0_LH p1_LH c_L p0_RH p1_RH c_R alpha error kappa

1 6 1029.7 0.294 0.706 1 0.706 0.294 0.000 0.877 0.294 0.4122 68 82.813 0.436 0.564 1 0.805 0.195 0.000 0.431 0.294 0.4123 33 0.992 0.129 0.871 1 0.603 0.397 0.000 0.493 0.268 0.4634 69 45.125 0.437 0.563 1 0.940 0.060 0.000 0.335 0.274 0.4525 37 63.938 0.356 0.644 1 0.603 0.397 0.000 0.462 0.271 0.4586 24 0.493 0.419 0.581 1 0.704 0.296 0.000 0.443 0.249 0.5037 43 22.930 0.448 0.552 1 0.654 0.346 0.000 0.407 0.254 0.4928 28 0.012 0.639 0.361 0 0.403 0.597 1.000 0.392 0.246 0.5089 23 1.369 0.496 0.504 1 0.961 0.039 0.000 0.111 0.246 0.50810 20 1.641 0.565 0.435 0 0.000 1.000 1.000 0.320 0.249 0.50311 23 1.369 0.449 0.551 1 0.957 0.043 0.000 0.276 0.240 0.52012 3 1.429 0.263 0.737 1 0.569 0.431 0.000 0.320 0.251 0.49713 69 45.125 0.452 0.548 1 0.924 0.076 0.000 0.252 0.240 0.52014 34 5.454 0.469 0.531 1 0.672 0.328 0.000 0.297 0.226 0.54815 31 7.552 0.529 0.471 0 0.842 0.158 0.000 0.246 0.226 0.54816 31 7.552 0.468 0.532 1 0.806 0.194 0.000 0.234 0.220 0.55917 32 8.937 0.572 0.428 0 0.228 0.772 1.000 0.348 0.215 0.57118 23 1.369 0.455 0.545 1 0.966 0.034 0.000 0.244 0.206 0.58819 20 0.836 0.615 0.385 0 0.442 0.558 1.000 0.352 0.223 0.55420 63 321.50 0.633 0.367 0 0.447 0.553 1.000 0.357 0.189 0.62121 68 77.250 0.488 0.512 1 0.727 0.273 0.000 0.212 0.203 0.59322 59 46.250 0.589 0.411 0 0.159 0.841 1.000 0.400 0.201 0.59923 23 1.369 0.463 0.537 1 0.954 0.046 0.000 0.209 0.189 0.62124 7 736.00 0.499 0.501 1 0.768 0.232 0.000 0.126 0.192 0.61625 42 8.750 0.363 0.637 1 0.591 0.409 0.000 0.362 0.189 0.62126 52 1.872 0.447 0.553 1 0.654 0.346 0.000 0.309 0.192 0.616

Page 34: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 21

27 61 -1.348 0.300 0.700 1 0.564 0.436 0.000 0.317 0.189 0.62128 57 278.81 0.450 0.550 1 0.747 0.253 0.000 0.266 0.178 0.64429 59 46.250 0.547 0.453 0 0.107 0.893 1.000 0.243 0.172 0.65530 25 0.092 0.868 0.132 0 0.457 0.543 1.000 0.223 0.167 0.66731 36 206.13 0.545 0.455 0 0.184 0.816 1.000 0.242 0.164 0.67232 47 23.103 0.696 0.304 0 0.443 0.557 1.000 0.290 0.172 0.65533 4 1.704 0.501 0.499 0 0.766 0.234 0.000 0.112 0.167 0.66734 4 1.704 0.474 0.526 1 0.745 0.255 0.000 0.186 0.175 0.65035 20 1.641 0.549 0.451 0 0.000 1.000 1.000 0.236 0.158 0.68436 23 1.369 0.466 0.534 1 0.946 0.054 0.000 0.186 0.169 0.66137 62 -0.869 0.262 0.738 1 0.548 0.452 0.000 0.254 0.150 0.70138 6 1279.9 0.455 0.545 1 0.812 0.188 0.000 0.233 0.164 0.67239 32 6.819 0.559 0.441 0 0.355 0.645 1.000 0.267 0.158 0.68440 32 0.707 0.305 0.695 1 0.520 0.480 0.000 0.094 0.164 0.67241 29 7.124 0.568 0.432 0 0.403 0.597 1.000 0.347 0.138 0.72342 12 2.000 0.218 0.782 1 0.521 0.479 0.000 0.194 0.138 0.72343 31 7.552 0.420 0.580 1 0.737 0.263 0.000 0.381 0.150 0.70144 33 0.992 0.183 0.817 1 0.546 0.454 0.000 0.245 0.133 0.73445 23 1.369 0.459 0.541 1 0.932 0.068 0.000 0.208 0.141 0.71846 55 975.56 0.236 0.764 1 0.541 0.459 0.000 0.227 0.136 0.72947 53 18.687 0.564 0.436 0 0.411 0.589 1.000 0.320 0.127 0.74648 35 249.00 0.371 0.629 1 0.573 0.427 0.000 0.391 0.130 0.74049 7 736.00 0.442 0.558 1 0.730 0.270 0.000 0.304 0.124 0.75150 48 60.067 0.451 0.549 1 0.606 0.394 0.000 0.319 0.138 0.723

Output 2.5 provides important information for the progress of the algorithm for eachiteration. Is shows that in the first 50 iterations, variable X23 is selected six times, whileX20, X31, and X32 are selected three times each; boosting is focusing on variables that areimportant for separating the data into classes. Also, a number of variables are not selectedin any iteration.

Figure 2.3 displays the error and kappa functions. Notice that after approximately 35iterations, the error levels off at approximately 0.15. At this point, AdaBoost is learningthe training data at a much slower rate.

Figure 2.3 Misclassification error (left panel) and kappa statistic (right panel) plots for the PERMY data setusing the %AdaBoost macro

Err

or

0.0

0.1

0.2

0.3

Iteration

1 11 21 31 41 51

Kap

pa

0.5

0.6

0.7

0.8

0.9

1.0

Iteration

1 11 21 31 41 51

Page 35: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

22 Pharmaceutical Statistics Using SAS: A Practical Guide

2.3.6 Implementation of Generalized Boosting AlgorithmsIn most model building applications, we need to evaluate the model’s performance on anindependent set of data. This process is referred to as validation or cross-validation. In thissection we provide a more general macro, %Boost, that can be used to evaluate bothtraining and testing data. This macro also implements the other generalized forms ofboosting.

The generalized boosting algorithms described in Section 2.3.1 can be implementedusing the same structure of the %AdaBoost macro by making several appropriateadjustments. To implement the Real AdaBoost algorithm, we must use predicted classprobabilities in place of predicted classification, and alter the observation contributions andweights. Because the %Split macro generates predicted class probabilities, we can directlyconstruct Real AdaBoost. However, both Gentle AdaBoost and LogitBoost require amethod that predicts a continuous, real-valued response. This stipulation requires that weimplement a different form of recursive partitioning. Instead of using partitioning based ona categorical response, we will implement partitioning based on a continuous response. Inshort, we seek to find the partition that minimizes the corrected sum-of-squares, ratherthan misclassification error or Gini Index, of the response across both new nodes. That is,we seek the variable, xj , and cut-point, c, that minimize∑

r1j

(y1j − y1j)2 +∑r2j

(y2j − y2j)2,

where r1j = {i : xj < c} and r2j = {i : xj ≥ c}.Like partitioning based on a categorical response, partitioning based on a continuous

response requires an exhaustive search to find the optimal cut-point. To find the variableand optimal cut-point, we have created a SAS/IML module, REGSPLIT IML. Thismodule is included in our comprehensive macro, %Boost, that implements the AdaBoost,Real AdaBoost, Gentle AdaBoost, and LogitBoost algorithms (the macro is provided onthe book’s companion Web site). The input variables for %Boost are the same as%AdaBoost. In addition, the user must specify the type of boosting to perform (TYPE=1performs AdaBoost, TYPE=2 performs Real AdaBoost, TYPE=3 performs GentleAdaBoost, and TYPE=4 performs LogitBoost). When AdaBoost or Real AdaBoost arerequested, %Boost produces an output data set with the same output variables as theAdaBoost macro. However, if Gentle AdaBoost or LogitBoost are specified, then a differentoutput data set is created. The variables included in this data set are listed below.

• ITER is the boosting iteration number.• REG VAR is the variable number that minimizes the corrected sum-of-squares.• MIN CSS is the minimum corrected sum-of-squares.• CUT VAL is the optimal cut-point.• YPRED L is the predicted value for observations less than CUT VAL.• YPRED R is the predicted value for observations greater than CUT VAL.• ERROR is the misclassification error of the cumulative boosting model at the current

iteration.• KAPPA is the kappa statistic of the cumulative boosting model at the current iteration.

As a diagnostic tool, the %Boost macro also creates an output data set namedOUTWTS that contains observation weights at the final boosting iteration. These can beused to identify observations that are difficult to classify.

To illustrate the %Boost macro, Program 2.6 analyzes the EXAMPLE1 data set usingthe Gentle AdaBoost algorithm with ten iterations.

Page 36: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 23

Program 2.6 Gentle AdaBoost algorithm in the recursive partitioning example

%boost(inputds=example1,p=2,outputds=out_ex1,outwts=out_wt1,iter=10,type=3);proc print data=out_ex1 noobs label;

format min_css cut_val ypred_l ypred_r error kappa 6.3;proc print data=out_wt1;

format weight 7.5;run;

Output from Program 2.6

OUT_EX1 data set

iter reg_var min_css cut_val ypred_L ypred_R error kappa

1 1 0.587 5.013 0.585 -0.702 0.180 0.6402 2 0.528 6.137 -0.574 0.835 0.160 0.6823 2 0.679 1.749 -1.000 0.357 0.050 0.9004 1 0.600 5.013 0.603 -0.656 0.070 0.8605 2 0.627 7.395 -0.388 1.000 0.000 1.0006 2 0.702 1.749 -1.000 0.365 0.000 1.0007 1 0.577 5.013 0.639 -0.659 0.000 1.0008 2 0.669 7.395 -0.343 1.000 0.000 1.0009 2 0.722 1.749 -1.000 0.343 0.000 1.000

10 1 0.566 5.013 0.651 -0.665 0.000 1.000

OUT_WT1 data set

Obs weight

1 0.000192 0.012073 0.033474 0.006095 0.018206 0.006097 0.000108 0.006099 0.0029510 0.00609

Output 2.6 provides a listing of the data set produced by the %Boost macro (OUT EX1data set). For the first iteration, X1 minimizes the corrected sum-of-squares (0.587) at thecut-point of 5.013. Observations that are less than this value have a predicted value of0.585, whereas observations greater than this value have a predicted value of −0.702. ForGentle AdaBoost, the error at the first iteration is 0.18, while the kappa value is 0.64.

In addition, Output 2.6 lists the first ten observations in the OUT WT1 data set, whichincludes the final weights for each observation after ten iterations. These weights areconstrained to sum to one; therefore, relatively high weights represent observations that aredifficult to classify, and observations with low weights are relatively easy to classify.

One can also model the permeability data using Gentle AdaBoost. But first we shouldsplit the data into training and testing sets in order to evaluate each model’s performanceon an independent set of data. For this chapter, the permeability data set has already beenseparated into training and testing sets through the SET variable. Program 2.7 creates thetraining and testing sets and invokes the %Boost macro to perform Gentle AdaBoost onthe training data with 35 iterations.

Page 37: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

24 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 2.7 Gentle AdaBoost algorithm in the permeability data example

data train;set permy(where=(set="TRAIN"));

data test;set permy(where=(set="TEST"));run;

%boost(inputds=train,p=71,outputds=genout1,outwts=genwt1,iter=35,type=3);proc print data=genout1 noobs label;

format min_css ypred_l ypred_r error kappa 6.3;run;

* Misclassification error;* Vertical axis;axis1 minor=none label=(angle=90 "Error") order=(0 to 0.3 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 36 by 5) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=genout1;

plot error*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

* Kappa statistic;* Vertical axis;axis1 minor=none label=(angle=90 "Kappa") order=(0.5 to 1 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 36 by 5) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=genout1;

plot kappa*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

Output from Program 2.7

iter reg_var min_css cut_val ypred_L ypred_R error kappa

1 6 0.838 1047.69 0.374 -0.434 0.299 0.4022 42 0.910 8.75 0.649 -0.142 0.294 0.4113 70 0.878 13.81 0.181 -0.734 0.238 0.5234 33 0.933 1.17 0.706 -0.116 0.234 0.5335 37 0.928 75.56 0.348 -0.201 0.238 0.5236 7 0.922 745.06 0.101 -0.803 0.234 0.5337 63 0.920 318.81 -0.325 0.241 0.196 0.6078 43 0.943 16.74 0.333 -0.172 0.178 0.6459 45 0.938 23.72 -0.464 0.136 0.173 0.65410 56 0.939 452.44 0.337 -0.186 0.192 0.61711 19 0.946 0.50 -0.349 0.157 0.154 0.69212 71 0.944 549.46 -0.078 0.751 0.131 0.73813 15 0.936 0.10 0.485 -0.122 0.126 0.74814 32 0.953 8.85 -0.057 0.716 0.126 0.74815 23 0.949 1.38 0.066 -0.924 0.112 0.77616 1 0.950 624.00 1.000 -0.065 0.112 0.77617 59 0.955 46.25 -0.037 0.849 0.103 0.79418 57 0.941 278.81 0.091 -0.738 0.107 0.78519 60 0.944 8.75 -0.142 0.424 0.098 0.80420 68 0.951 84.94 0.073 -0.704 0.107 0.78521 34 0.955 0.71 -0.539 0.074 0.103 0.794

Page 38: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 25

22 41 0.950 9.88 0.747 -0.137 0.103 0.79423 34 0.956 0.71 -0.518 0.095 0.112 0.77624 33 0.949 0.99 0.724 -0.137 0.107 0.78525 13 0.957 0.02 1.000 -0.036 0.103 0.79426 57 0.959 274.63 0.085 -0.573 0.103 0.79427 32 0.957 1.58 0.191 -0.224 0.079 0.84128 34 0.947 0.71 -0.584 0.093 0.084 0.83229 32 0.938 8.85 -0.148 0.856 0.084 0.83230 34 0.951 0.71 -0.546 0.099 0.070 0.86031 34 0.947 0.71 -0.612 0.000 0.070 0.86032 34 0.947 0.71 -0.612 0.000 0.070 0.86033 34 0.947 0.71 -0.612 -0.000 0.070 0.86034 34 0.947 0.71 -0.612 -0.000 0.070 0.86035 34 0.947 0.71 -0.612 -0.000 0.070 0.860

Output 2.7 lists the GENOUT1 data set created by the %Boost macro. Figure 2.4illustrates the performance of the Gentle AdaBoost algorithm on the training data set.Notice that the training error decreases from 0.3 to around 0.1 after approximately 25iterations.

Figure 2.4 Misclassification error (left panel) and kappa statistic (right panel) plots for the training subset ofthe PERMY data set using the %Boost macro (Gentle AdaBoost algorithm)

Err

or

0.0

0.1

0.2

0.3

Iteration

1 6 11 16 21 26 31 36

Kap

pa

0.5

0.6

0.7

0.8

0.9

1.0

Iteration

1 6 11 16 21 26 31 36

%Predict MacroTo complement the %Boost macro, we have also developed a macro, %Predict, to predictnew observations using the model information generated by %Boost. This macro can beused to evaluate the performance of a model on an independent testing or validation dataset. The input variables to %Predict are:

• PRED DS is the name of the data set to be predicted. This data set must havedescriptors named X1, X2, . . . , XP. The response must be named Y and must assumevalues of 0 or 1.

• P is the number of descriptors in the input data set.• BOOST DS is the name of the data set with boost model information (the OUTPUTDS

from the %Boost macro).• TYPE is the type of boosting for prediction.• ITER is the number of boosting iterations desired.

Page 39: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

26 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 2.8 calls the %Predict macro to examine the predictive properties of theclassification model in the permeability data example. The program analyzes the TESTdata set created in Program 2.7.

Program 2.8 Prediction in the permeability data example

%predict(pred_ds=test,p=71,boost_ds=genout1,outputds=gentst1,out_pred=genpred1,type=3,iter=35);

* Misclassification error;* Vertical axis;axis1 minor=none label=(angle=90 "Error") order=(0 to 0.5 by 0.1) width=1;* Horizontal axis;axis2 minor=none label=("Iteration") order=(1 to 36 by 5) width=1;symbol1 i=join value=none color=black width=5;proc gplot data=gentst1;

plot error*iter/vaxis=axis1 haxis=axis2 frame;run;quit;

Figure 2.5 depicts changes in the model error across iterations. Although we saw adecrease in the misclassification error for the training set across iterations (Figure 2.4), thesame is not true for the test set. Instead, the misclassification error goes up slightly acrossiterations. For drug discovery data, this phenomenon is not surprising—often the responsein many data sets is noisy (a number of samples have been misclassified). Thesemisclassified samples hamper the learning ability of many models, preventing them fromlearning the correct structure between the descriptors and the classification variable. InSection 2.4, we further explain how to use boosting to identify noisy or mislabeledobservations. Removing these observations often produces a better overall predictive model.

Figure 2.5 Misclassification rates in the testing subset of the PERMY data set using the %Predict macro(Gentle AdaBoost algorithm)

Err

or

0.0

0.1

0.2

0.3

0.4

0.5

Iteration

1 6 11 16 21 26 31 36

2.3.7 Properties of BoostingMany authors have shown that boosting is generally robust to overfitting; that is, as thecomplexity of the boosting model increases for the training set (i.e., the number ofiterations increases), the test set error does not increase (Schapire, Freund, Bartlett andLee, 1998; Friedman, Hastie, and Tibshirani, 2000; Freund, Mansour and Schapire, 2001).

Page 40: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 27

However, boosting, by its inherent construction, is vulnerable to certain types of dataproblems, which can make it difficult for boosting to find a model that can be generalized.

The primary known problem for boosting is noise in the response (Krieger, Long, andWyner, 2002; Schapire, 2002; Optiz and Maclin, 1999), which can occur when observationsare mislabeled or classes overlap in descriptor space. Under either of these circumstances,boosting will place increasing higher weights on those observations that are inconsistentwith the majority of the data. As a check-valve, the performance of the model at each stageis evaluated against the original training set. Models that poorly classify receive lessweight, while models that accurately classify receive more weight. But, the finalagglomerative model has attempted to fit the noise in the response. Hence, the final modelwill use these rules to predict new observations, thus increasing prediction error.

For similar reasons, boosting will be negatively affected by observations that are outliersin the descriptor space. In fact, because of this problem, Freund and Schapire (2001)recommend removing known outliers before performing boosting.

Although noise can be the Achilles heel of boosting, its internal correction methods canhelp us identify potentially mislabeled or outlying observations. Specifically, as thealgorithm iterates, boosting will place increasing larger weights on observations that aredifficult to classify. Hence, tracking observation weights can be used as a diagnostic tool foridentifying these problematic observations. In the following section, we will illustrate theimportance of examining observation weights.

2.4 Model BuildingThis section discusses model building with the %Boost macro introduced in Section 2.3.

Each modification of the AdaBoost algorithm (AdaBoost, Real AdaBoost, GentleAdaBoost, and LogitBoost) performs differently on various types of data. Because of thisfact, we should evaluate the performance of each model on a training and testing set toidentify the model with the best performance. For the permeability data, Program 2.9performs each version of boosting on the training and testing sets (TRAIN and TEST datasets) created in Program 2.7. The program also computes and plots the associatedmisclassification errors to gauge the predictive ability of each model.

Program 2.9 Performance of the four boosting algorithms in the permeability data example

* AdaBoost;%boost(inputds=train,p=71,outputds=adaout1,outwts=adawt1,iter=40,type=1);%predict(pred_ds=test,p=71,boost_ds=adaout1,outputds=adatst1,out_pred=adapred1,

type=1,iter=40);* Real AdaBoost;%boost(inputds=train,p=71,outputds=realout1,outwts=realwt1,iter=40,type=2);%predict(pred_ds=test,p=71,boost_ds=realout1,outputds=realtst1,out_pred=realpred1,

type=2,iter=40);* Gentle AdaBoost;%boost(inputds=train,p=71,outputds=genout1,outwts=genwt1,iter=40,type=3);%predict(pred_ds=test,p=71,boost_ds=genout1,outputds=gentst1,out_pred=genprd1,

type=3,iter=40);* LogitBoost;%boost(inputds=train,p=71,outputds=logout1,outwts=logwt1,iter=40,type=4);%predict(pred_ds=test,p=71,boost_ds=logout1,outputds=logtst1,out_pred=logprd1,

type=4,iter=40);* Training set misclassification errors;data adaout1;

set adaout1; method=1;

Page 41: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

28 Pharmaceutical Statistics Using SAS: A Practical Guide

data realout1;set realout1; method=2;

data genout1;set genout1; method=3;

data logout1;set logout1; method=4;

data train_error;set adaout1(keep=iter error method)realout1(keep=iter error method)genout1(keep=iter error method)logout1(keep=iter error method);

axis1 minor=none label=(angle=90 "Error") order=(0 to 0.3 by 0.1) width=1;axis2 minor=none label=("Iteration") order=(1 to 41 by 10) width=1;symbol1 i=join value=none color=black line=1 width=3;symbol2 i=join value=none color=black line=34 width=3;symbol3 i=join value=none color=black line=20 width=3;symbol4 i=join value=none color=black line=41 width=3;proc gplot data=train_error;

plot error*iter=method/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

* Test set misclassification errors;data adatst1;

set adatst1; method=1;data realtst1;

set realtst1; method=2;data gentst1;

set gentst1; method=3;data logtst1;

set logtst1; method=4;data test_error;

set adatst1(keep=iter error method)realtst1(keep=iter error method)gentst1(keep=iter error method)logtst1(keep=iter error method);

axis1 minor=none label=(angle=90 "Error") order=(0.2 to 0.5 by 0.1) width=1;axis2 minor=none label=("Iteration") order=(1 to 41 by 10) width=1;symbol1 i=join value=none color=black line=1 width=3;symbol2 i=join value=none color=black line=34 width=3;symbol3 i=join value=none color=black line=20 width=3;symbol4 i=join value=none color=black line=41 width=3;proc gplot data=test_error;

plot error*iter=method/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

The misclassification error curves for the four boosting algorithms are displayed in theleft panel of Figure 2.6. After 40 iterations, each method has learned as much informationas possible about the structure of the training set in relation to compound permeabilityclassification (notice the flattening of each misclassification curve). These models aresubsequently applied to the test set and the resulting misclassification error curves areshown in the right panel of Figure 2.6. This plot demonstrates that the predictive ability ofboosting does not improve as the number of iterations increases. Notice, however, thatboosting does not rapidly overfit; the test set error does not climb rapidly as iterationnumber increases.

Page 42: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 29

Figure 2.6 Misclassification rates the training (left panel) and test (right panel) subsets of the PERMY dataset (solid curve, AdaBoost; dotted curve, Real AdaBoost; dashed curve, Gentle AdaBoost; dashed-dotted curve,LogitBoost)

Err

or

0.0

0.1

0.2

0.3

Iteration

1 11 21 31 41

Err

or

0.2

0.3

0.4

0.5

Iteration

1 11 21 31 41

As mentioned in the previous section, boosting will focus its modeling efforts onobservations that are difficult to classify by increasing the observations’ weights. Theoriginal classification of compounds in this data set is inherently noisy, which could becausing boosting to focus on incorrect features of the data in an attempt to classify allobservations. Program 2.10 computes and plots the Real AdaBoost weights of eachobservation after 40 iterations.

Program 2.10 Observation weights for the Real AdaBoost algorithm

%boost(inputds=train,p=71,outputds=realout1,outwts=realwt1,iter=40,type=2);* Weights from Real AdaBoost;proc sort data=realwt1 out=sortwts;

by descending weight;data sortwts;

set sortwts;obsnum=_n_;

* Vertical axis;axis1 minor=none label=(angle=90 "Weight") order=(0 to 0.03 by 0.01) width=1;* Horizontal axis;axis2 minor=none label=("Observations") order=(0 to 225 by 25) width=1;symbol1 i=none value=circle color=black height=4;proc gplot data=sortwts;

plot weight*obsnum/vaxis=axis1 haxis=axis2 frame;run;quit;

Figure 2.7 displays the weights computed by Program 2.10. In this example, the RealAdaBoost algorithm focuses 50 percent of the weight on only 20 percent of the data—atelltale sign that boosting is chasing misclassified or difficult-to-classify observations.

In an attempt to find a predictive model, we have removed these observations and havere-run each boosting algorithm (Program 2.11). The misclassification error curvesgenerated by Program 2.11 are displayed in Figure 2.8.

Page 43: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

30 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 2.7 Observation weights for the Real AdaBoost algorithm after 40 iterations (observations are orderedby magnitude)

Wei

ght

0.00

0.01

0.02

0.03

Observations

0 25 50 75 100 125 150 175 200 225

Program 2.11 Performance of the four boosting algorithms in the permeability data example (reduced dataset)

* Real AdaBoost;%boost(inputds=train,p=71,outputds=realout1,outwts=realwt1,iter=40,type=2);data newtrain;

merge train realwt1;if weight>0.007 then delete;drop weight;

* AdaBoost;%boost(inputds=newtrain,p=71,outputds=adaout2,outwts=adawt2,iter=40,type=1);%predict(pred_ds=test,p=71,boost_ds=adaout2,outputds=adatst2,out_pred=adapred2,

type=1,iter=40);* Real AdaBoost;%boost(inputds=newtrain,p=71,outputds=realout2,outwts=realwt2,iter=40,type=2);%predict(pred_ds=test,p=71,boost_ds=realout2,outputds=realtst2,out_pred=realpred2,

type=2,iter=40);* Gentle AdaBoost;%boost(inputds=newtrain,p=71,outputds=genout2,outwts=genwt2,iter=40,type=3);%predict(pred_ds=test,p=71,boost_ds=genout2,outputds=gentst2,out_pred=genprd2,

type=3,iter=40);* LogitBoost;%boost(inputds=newtrain,p=71,outputds=logout2,outwts=logwt2,iter=40,type=4);%predict(pred_ds=test,p=71,boost_ds=logout2,outputds=logtst2,out_pred=logprd2,

type=4,iter=40);* Training set misclassification errors;data adaout2;

set adaout2; method=1;data realout2;

set realout2; method=2;data genout2;

set genout2; method=3;data logout2;

set logout2; method=4;

Page 44: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 31

data train_error2;set adaout2(keep=iter error method)realout2(keep=iter error method)genout2(keep=iter error method)logout2(keep=iter error method);

axis1 minor=none label=(angle=90 "Error") order=(0 to 0.3 by 0.1) width=1;axis2 minor=none label=("Iteration") order=(1 to 41 by 10) width=1;symbol1 i=join value=none color=black line=1 width=3;symbol2 i=join value=none color=black line=34 width=3;symbol3 i=join value=none color=black line=20 width=3;symbol4 i=join value=none color=black line=41 width=3;proc gplot data=train_error2;

plot error*iter=method/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

* Test set misclassification errors;data adatst2;

set adatst2; method=1;data realtst2;

set realtst2; method=2;data gentst2;

set gentst2; method=3;data logtst2;

set logtst2; method=4; run;data test_error2;

set adatst2(keep=iter error method)realtst2(keep=iter error method)gentst2(keep=iter error method)logtst2(keep=iter error method);

axis1 minor=none label=(angle=90 "Error") order=(0.2 to 0.5 by 0.1) width=1;axis2 minor=none label=("Iteration") order=(1 to 41 by 10) width=1;symbol1 i=join value=none color=black line=1 width=3;symbol2 i=join value=none color=black line=34 width=3;symbol3 i=join value=none color=black line=20 width=3;symbol4 i=join value=none color=black line=41 width=3;proc gplot data=test_error2;

plot error*iter=method/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

Figure 2.8 depicts the misclassification error curves for the four boosting algorithms inthe reduced data set. The initial training error is lower than for the original training set(see Figure 2.6), and each boosting method more rapidly learns the features of the trainingdata that are related to permeability classification. This implies that the observations thatwere removed from the training set were likely misclassified, but the predictive performanceon the test set does not improve (see the right panel of Figure 2.8).

Undoubtedly, the test set also contains observations that are misclassified. In a finalattempt to improve boosting’s predictive ability, we have run Real AdaBoost on the testset and have removed the highest weighted observations. To this reduced test set, we haveapplied each reduced training set boosting model (Program 2.12). The misclassificationerror rates computed by the program are shown in Figure 2.9.

Page 45: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

32 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 2.8 Misclassification rates in the training (left panel) and test (right panel) subsets for the reducedtraining set (solid curve, AdaBoost; dotted curve, Real AdaBoost; dashed curve, Gentle AdaBoost; dashed-dotted curve, LogitBoost)

Err

or

0.0

0.1

0.2

0.3

Iteration

1 11 21 31 41

Err

or

0.2

0.3

0.4

0.5

Iteration

1 11 21 31 41

Program 2.12 Predictive performance of the four boosting algorithms in the permeability data example(reduced data set)

* Model test set data and identify compounds with high weights;%boost(inputds=test,p=71,outputds=realout3,outwts=realwt3,iter=40,type=1);data newtest;

merge test realwt3;if weight>0.01 then delete;drop weight;

* AdaBoost;%predict(pred_ds=newtest,p=71,boost_ds=adaout2,outputds=adatst3,out_pred=adapred3,

type=1,iter=40);* Real AdaBoost;%predict(pred_ds=newtest,p=71,boost_ds=realout2,outputds=realtst3,out_pred=realpred3,

type=2,iter=40);* Gentle AdaBoost;%predict(pred_ds=newtest,p=71,boost_ds=genout2,outputds=gentst3,out_pred=genprd3,

type=3,iter=40);* LogitBoost;%predict(pred_ds=newtest,p=71,boost_ds=logout2,outputds=logtst3,out_pred=logprd3,

type=4,iter=40);* Test set misclassification errors;data adatst3;

set adatst3; method=1;data realtst3;

set realtst3; method=2;data gentst3;

set gentst3; method=3;data logtst3;

set logtst3; method=4;data test_error3;

set adatst3(keep=iter error method)realtst3(keep=iter error method)gentst3(keep=iter error method)logtst3(keep=iter error method);

Page 46: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 33

axis1 minor=none label=(angle=90 "Error") order=(0 to 0.3 by 0.1) width=1;axis2 minor=none label=("Iteration") order=(1 to 41 by 10) width=1;symbol1 i=join value=none color=black line=1 width=3;symbol2 i=join value=none color=black line=34 width=3;symbol3 i=join value=none color=black line=20 width=3;symbol4 i=join value=none color=black line=41 width=3;proc gplot data=test_error3;

plot error*iter=method/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

Figure 2.9 Misclassification error rates in the test subset of the reduced set (solid curve, AdaBoost; dottedcurve, Real AdaBoost; dashed curve, Gentle AdaBoost; dashed-dotted curve, LogitBoost)

Err

or

0.0

0.1

0.2

0.3

Iteration

1 11 21 31 41

Figure 2.9 demonstrates that the misclassification error rates on this set are noticeablylower than on the original test set.

While boosting does not significantly reduce test set classification error across iterationsfor this example, it does allow the user to identify difficult-to-classify, or possiblymisclassified observations. With these observations present, any method will have a difficulttime finding the underlying relationship between predictors and response.

2.5 Partial Least Squares for DiscriminationPerhaps the most commonly used method of statistical discrimination is simple lineardiscriminant analysis (LDA). A fundamental problem associated with the use of LDA inpractice, however, is that the associated pooled within-groups sums-of-squares andcross-products matrix has to be invertible. For data sets with collinear features or withsignificantly more features than observations this may not be the case. In these situationsthere are many different options available. By far the most common in the practice ofchemometrics is to first use principal components analysis (PCA) to reduce the dimensionof the data, and then to follow the PCA with a version of LDA, or to simply impose an adhoc classification rule at the level of the PCA scores and leave it at that. While bothapproaches are sometimes successful in identifying class structure, the dimension reductionstep was not focused on the ultimate goal of discrimination.

Of course, PCA is not the only option for collinear data. Ridging or “shrinkage” can beemployed to stabilize the pertinent covariance matrices so that the classical discriminationparadigms might be implemented (Friedman, 1989; Rayens, 1990; Rayens and Greene,

Page 47: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

34 Pharmaceutical Statistics Using SAS: A Practical Guide

1991; Greene and Rayens, 1989). Alternatively, variable selection routines based in geneticalgorithms are gaining popularity (e.g., Lavine, Davidson and Rayens, 2004) and have beenshown to be successful in particular on microarray data. Other popular methods includeflexible discriminant analysis (Ripley, 1996) and penalized discriminant analysis (Hastieet al., 1995), which are also variations on the ridging theme.

It is well known that PCA is not a particularly reliable paradigm for discriminationsince, unlike simple LDA, it is capable of identifying only gross variability and is notcapable of distinguishing “among-groups” and “within-groups” variables. Indeed, a PCAapproach to discrimination is typically successful only when the among-groups variabilitydominates the within-groups variability, as often happens with chromatography studies.This point is illustrated in a small simulation study by Barker and Rayens (2003).

It turns out that partial least squares (PLS) is much more appropriate than PCA as adimension-reducing technique for the purposes of linear discrimination. Barker and Rayensfully established this connection between PLS and LDA, and we will refer to their use ofPLS for facilitating a discriminant analysis as PLS-LDA. Some of the details of thisconnection will be reviewed before illustrating how PLS-LDA can easily be implemented inSAS using the PLS procedure.

To facilitate the discussion, we will use PROC PLS to perform PLS-LDA on the samepermeability data set that we used to illustrate boosting.

2.5.1 Review of PLSPLS is based on Herman Wold’s original Nonlinear Iterative Partial Least Squares(NIPALS) algorithm (Wold, 1966; Wold, 1981), adapted to reduce dimensionality in theoverdetermined regression problem. However, there are many different versions of the sameparadigm, some of which allow the original PLS problem to be viewed as a collection ofwell-posed eigenstructure problems which better facilitate PLS being connected tocanonical covariates analysis (CCA) and, ultimately, to LDA. This is the perspective takenon PLS in this chapter. With respect to SAS, this perspective is essentially equivalent toinvoking the METHOD=SIMPLS option, which we will revisit in the discussion below.

It should be noted at the outset that under this general eigenstructure perspective it isespecially obvious that different sets of constraints—and whether these constraints areimposed in both the X- and Y -spaces or only one—will lead to different PLS directions.This does not seem to be well known, or at least not often discussed in practice. With PCAthe resulting “directions” or weight vectors are orthogonal and, at the same time, theassociated component scores are uncorrelated. With PLS one has to choose since bothuncorrelated score constraints and orthogonal weights are not simultaneously possible.Which constraint seems to be important and where you impose them depends on who youtalk to. In the neurosciences, orthogonal constraints are considered essential, since thedirections are often more important than the scores and having the directions beorthogonal (interpreted in practice as “independent”) helps with the interpretation of theestimated “signals”. Chemometricians, however, typically have no interest in an orthogonaldecomposition of variable space and are much more focused on creating “new” data withuncorrelated features, which makes sense, since it is often a problem of collinearity thatbrings them to PLS in the first place.

For example, this latter perspective is adopted in PROC PLS where uncorrelated scoreconstraints are implicitly assumed. We are going to simplify our presentation by largelyignoring the constraints issue. Indeed, with all the standard constraint sets the product ofthe sample covariance matrix and its transpose, SxySyx, emerges at the core construct inthe extraction of the PLS structure. As we will see below, it is this construct that isintimately related to Fisher’s LDA. First, the definition of PLS is reviewed.

Page 48: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 35

Definition of PLSLet x and y be random p- and q-dimensional vectors with dispersion matrices Σx and Σy,respectively (the sample matrices will be denoted by Sx and Sy). Denote the covariance ofx and y by Σxy. The first pair of PLS directions are defined as p- and q-dimensional vectorsa and b that jointly maximize

Cov[(aT x, bT y)]2

(aT a)(bT b).

The objective of the maximization in this definition can be more clearly seen by rewritingit as

Cov[(aT x, bT y)]2

(aT a)(bT b)= Var(aT x)[Corr(aT x, bT y)]2Var(bT y).

In this framework, it is easy to see how PLS can be thought of as “penalized” canonicalcorrelations analysis (see Frank and Friedman, 1993). That is, the squared correlation termalone represents the objective of CCA. However, this objective is penalized by a varianceterm for the X-space and another for the Y -space. These variance terms represent theobjective of principal components analysis. Hence, from this representation, PLS seeks tomaximize correlation while simultaneously reducing dimension in the X- and Y -spaces.

PLS TheoremThe first PLS direction in the X-space, a1, is an eigenvector of ΣxyΣyx corresponding to thelargest eigenvalue, and the corresponding first PLS direction in the Y -space, b1, is given byb1 = Σyxa1.

If orthogonality constraints are imposed on the directions in both the X- and Y -spaces,then any subsequent PLS solution, say ak+1, follows analogously, with the computation ofthe eigenvector of SxySyx corresponding to the (k + 1)st largest eigenvalue and the Y -spacedirection emerges as bk+1 = Syxak+!. If, instead of orthogonal constraints, uncorrelated scoreconstraints are imposed, then the (k + 1)st X-space direction is an eigenvectorcorresponding to the largest eigenvalue of

(Ip − (ΣxA(k))

[(ΣxA

(k))T ΣxA(k)

]−1(ΣxA

(k))T )ΣxyΣyx,

where A(k) = [a1, a2, . . . , ak]p×k. There is a completely similar expression for the Y -spacestructure.

The described theorem is well known in the PLS literature and, for uncorrelated scoreconstraints, a variant was first produced by de Jong (1993) when the SIMPLS procedurewas introduced. Another proof of this Theorem under different constraint sets (and novel inthat it does not use Lagrange multipliers) is provided in the technical report by Rayens(2000). For now, the point is simply that the derivation of the PLS directions will eitherdirectly or indirectly involve the eigenstructure of SxySyx.

In the following presentation, CCA is first connected to LDA, then, using thisconnection between PLS and CCA, PLS is formally connected and finally, PLS to LDA.

2.5.2 Connection between CCA and LDAWhen CCA is performed on a training set, X (e.g., 71 molecular properties measured on354 compounds described in Section 2.2) and an indicator matrix, Y , representing groupmembership (e.g., a 1 for a permeable compound and a 0 for a non-permeable compound),the CCA directions are just Fisher’s LDA directions. This well-known fact, which was firstrecognized by Bartlett (1938), has been reproved more elegantly by Barker and Rayens(2003) by using the following results, which are important for this presentation.

Page 49: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

36 Pharmaceutical Statistics Using SAS: A Practical Guide

Let xij be the p-dimensional vector for the jth observation in the ith group and g be thenumber of groups. Denote the training set consisting of ni observations on each of p featurevariables by

Xn×p = (x11, x12, . . . , x1n1 , . . . , xg1, xg2, . . . , xgng)T .

Let H denote the among-groups sums-of-squares and cross-products matrix and let E bethe pooled within-groups sums-of-squares and cross-products matrix,

H =g∑

i=1

ni(xi − x)(xi − x)T , E =g∑

i=1

ni∑j=1

(xij − xi)(xij − xi)T ,

where

xi = (1/ni)ni∑

j=1

xij , x = (1/n)g∑

i=1

ni∑j=1

xij , n =g∑

i=1

ni.

LDA, also known as “canonical discriminant analysis” (CDA) when expressed this way,manipulates the eigenstructure of E−1H. The connections between CDA and a perspectiveon LDA that is more focused on the minimization of misclassification probabilities arewell-known and will not be repeated here (see Kshiragar and Arseven, 1975).

There are two obvious ways that one can code the group membership in the matrix Y ,either

Y =

⎡⎢⎢⎢⎣1n1×1 0 · · · 0

0 1n2×1 · · · 0...

.... . .

...0 0 · · · 1ng×1

⎤⎥⎥⎥⎦ or Z =

⎡⎢⎢⎢⎢⎢⎣1n1×1 0 · · · 0

0 1n2×1 · · · 0...

.... . .

...0 0 · · · 1ng−1×10 0 · · · 0

⎤⎥⎥⎥⎥⎥⎦ .

For example, for the permeability data one could rationally choose

Y =[

1177×1 0177×10177×1 1177×1

]or Z =

[1177×10177×1

].

Note that the sample “covariance” matrix Sy is g × g and rank g − 1, while Sz is(g − 1) × (g − 1) and rank (g − 1). Regardless, the fact that both Y and Z are indicatormatrices suggests that Sy and Sz will have special forms. Barker and Rayens (2003) showedthe following:

S−1z = (n − 1)

(1ng

1g−11Tg−1 + M−1

),

where

M =

⎡⎢⎢⎢⎣n1 0 · · · 00 n2 · · · 0...

.... . .

...0 0 · · · ng−1

⎤⎥⎥⎥⎦(g−1)×(g−1)

, Scy =

[S−1

z 0g−10g−1 0

]g×g

.

With these simple expressions for S−1z and Sc

y, it is possible to relate constructs that areessential to PLS-LDA to Fisher’s H matrix, as will be seen in the next section.

Page 50: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 37

2.5.3 Connection between PLS and LDASince PLS is just a penalized version of CCA and CCA is, in turn, related to LDA(Bartlett, 1938), it is reasonable to expect that PLS might have some direct connection toLDA. Barker and Rayens (2003) gave a formal statistical explanation of this connection. Inparticular, recall that above the USC-PLS directions were associated with theeigenstructure of SxySyx or, for a classification application, SxzSzx, depending on how themembership matrix was coded. In either case, Barker and Rayens (2003) related thepertinent eigenstructure problem to H as follows:

SxySyx = H∗ =1

n − 1

g∑i=1

n2i (xi − x)(xi − x)T ,

SxzSzx = H∗∗ =1

n − 1

g−1∑i=1

n2i (xi − x)(xi − x)T .

It should be noted that when the Y block is coded with dummy variables, the presence ofthe Y -space penalty (Var(bT y)) in the definition of PLS does not seem to be all thatappropriate, since Y -space variability is not meaningful. Barker and Rayens (2003)removed this Y -space penalty from the original objective function, reposed the PLSoptimization problem, and were able to show that the essential eigenstructure problem thatis being manipulated in this case is one that involves exactly H, and not merely H∗ or H∗∗.However, in the case of two groups, as with our permeability data, it is not hard to showthat

H∗ = 2H∗∗ =n − 1

2

(1n1

+1n2

)H

and, hence, that the eigenstructure problems are equivalent and all proportional to(x1 − x)(x1 − x)T .

The practical upshot is simple: when one uses PLS to facilitate discrimination in the“obvious” way, with the intuition of “predicting” group membership from a training set,then the attending PLS eigenstructure problem is one that depends on (essentially)Fisher’s among-groups sums-of-squares and cross-products matrix H. It is, therefore, nosurprise that PLS should perform better than PCA for dimension reduction whendiscriminant analysis on the scores is the ultimate goal. This is simply because thedimension reduction provided by PLS is determined by among-groups variability, while thedimension reduction provided by PCA is determined by total variability. It is easy toimplement PLS-LDA in SAS, and this is discussed next.

2.5.4 Logic of PLS-LDAThere are many rational ways that PLS can be used to facilitate discrimination. Consistentwith how PCA has been used for this purpose, some will choose to simply plot the first twoor three PLS-LDA “scores” and visually inspect the degree of separation, a process that isentirely consistent with the spirit of “territory plots” and classical CDA. The classificationof an unknown may take place informally, say, by mapping it to the same two or threedimensional space occupied by the scores and then assigning it to the group that admitsthe closest mean score, where “closest” may be assessed in terms of Mahalanobis distanceor Euclidean distance.

Alternately, the scores themselves may be viewed simply as new data that have beenappropriately “prepared” for an LDA, and a full LDA might be performed on the scores,either with misclassification rates as a goal (the DISCRIM procedure) or visual separationand territory plots as a goal (the CANDISC procedure). Figure 2.10 helps illustrate themany options. In Section 2.5.5, we will discuss how to produce the PLS-LDA scores withthe PLS procedure.

Page 51: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

38 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 2.10 Schematic depicting the logic of PLS-LDA

Training data Xn×p

(choose q PLS components)

PLS-LDA scores Wn×q

� �

Plot scores,look for separation(classifying using

ad hoc rule)

Send to formalclassification procedure,

PROC CANDISC or PROC DISCRIM(classifying using formal rule)

� �

Find correspondingscore wq×1

Unknown xp×1

2.5.5 ApplicationAs mentioned in the Introduction, drug discovery teams often generate data that containboth descriptor information and a categorical measurement. Often, the teams desire tosearch for a model that can reasonably predict the response. For this section, we applyPLS-LDA to the permeability data set (the PERMY data set can be found on the book’scompanion Web site) introduced in Section 2.2.

Recall that this data set contains 354 compounds that have been screened to assesspermeability. While the original measure of permeability was a continuous value, scientistshave categorized these compounds into groups of permeable and non-permeable. Inaddition, this data set contains an equal number of compounds in each group. Usingin-house software, 71 molecular properties, thought to be related to compoundpermeability, were generated for each compound and were used as the descriptors for bothmodeling techniques.

This is not an uncommon drug discovery-type data set—the original continuousresponse is noisy and is subsequently reduced to a binary response. Hence, we fully expectsome compounds to be incorrectly classified. Also, the descriptor set is computationallyderived and is over-described. With these inherent problems, many techniques would havea difficult time finding a relationship between the descriptors and the response.

Implementing PLS-LDA in SAS

The purpose of this presentation is not to detail the many options that are well known inPROC DISCRIM or PROC CANDISC, or perhaps less well known in PROC PLS. Rather,this presentation is focused on simply showing the reader how to use PROC PLS inconjunction with, say, PROC DISCRIM to implement PLS-LDA as described above. Tobetter facilitate this discussion,f we will use the permeability data, as described in the

Page 52: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 39

previous subsection. For this analysis, all observations are used in the training set andcross-validation is used to assess model performance.

Notice with subgroup matrices of size 177 by 71 one would not necessarily expect thatany dimension reduction would be necessary and that PROC DISCRIM could be applieddirectly to the training set. However, many of the descriptors are physically related, so, weexpect these descriptors to be correlated to some extent. In fact, each of the individualgroup covariance matrices, as well as the pooled covariance matrix, are 71 by 71 but onlyhave rank 63. So, in theory, Fisher’s linear discriminant analysis—whether in the form thatfocuses on misclassification probabilities (PROC DISCRIM) or visual among-groupsseparation (PROC CANDISC)—cannot be applied, owing to the singularity of thesematrices. Hence, these data are good candidates for first reducing dimension and thenperforming a formal discriminant analysis.

There is an alternative that has to be mentioned, however. When confronted with asingular covariance matrix, PROC DISCRIM will issue an innocuous warning and thenproceed to construct a classification rule anyway. It is not widely known, perhaps, but inthis situation SAS will employ a so-called “quasi-inverse,” which is a SAS innovation. Toconstruct the quasi-inverse “small values” are arbitrarily added to zero variance directions,thereby forcing the corresponding covariance matrices to be nonsingular and preserving thebasic rationale behind the classification paradigm. Again, the purpose of this illustration isnot to compare dimension reduction followed by classification with direction classificationusing a quasi-inverse, but it is important that the reader be aware that SAS already hasone method of dealing with collinear discriminant features.

Program 2.13 performs the PLS-LDA analysis of the permeability data using PROCPLS. The initial PROC DISCRIM is invoked solely to produce the results of using thequasi-inverse. Keep in mind that the Y variable is a binary variable in this two-group case,designed to reflect corresponding group membership. In this example the first ten PLSscores (XSCR1-XSCR10) are used for the discrimination (the LV=10 option is used torequest ten PLS components). For clarity of comparison, the pooled covariance matrix wasused in all cases.

Program 2.13 PLS-LDA analysis of the permeability data

ods listing close;proc discrim data=permy list crosslist noprint;

class y;var x1-x71;run;

ods listing;proc pls data=permy method=simpls lv=10;

model y=x1-x71;output out=outpls xscore=xscr;

proc print data=outpls;var xscr1 xscr2;run;

ods listing close;proc discrim data=outpls list crosslist noprint;

class y;var xscr1-xscr10;run;

ods listing;

Page 53: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

40 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 2.13

The PLS Procedure

Percent Variation Accounted for by SIMPLS Factors

Number ofExtracted Model Effects Dependent VariablesFactors Current Total Current Total

1 26.6959 26.6959 18.5081 18.50812 16.6652 43.3611 2.9840 21.49213 8.3484 51.7096 3.0567 24.54884 3.5529 55.2625 3.2027 27.75155 6.2873 61.5498 0.8357 28.58726 4.0858 65.6356 0.8749 29.46217 3.5761 69.2117 0.8217 30.28388 7.0819 76.2935 0.4232 30.70709 2.2274 78.5209 0.9629 31.6699

10 4.7272 83.2481 0.4038 32.0737

Obs xscr1 xscr2

1 -4.2733 -0.786082 -3.7668 -5.290113 2.0578 8.044454 -1.0728 -0.523245 -5.0157 2.379406 1.7600 -0.930717 -5.2317 3.229518 -4.2919 -6.718169 -6.3754 -3.81705

10 0.8542 -0.96708

Output 2.13 displays a “variance summary” produced by PROC PLS as well as a listingof first ten values of the two PLS components (XSCR1 and XSCR2). In general, navigatingand understanding the PROC PLS output was discussed in great detail by Tobias (1997a,1997b), and there is little need for us to repeat that discussion here. Rather, our purpose issimply to extract and then access the PLS scores for purposes of performing either a formalor ad hoc classification analysis. However, it is relevant to see how well the original featurespace has been summarized by these extracted components.

When ten PLS components are requested, the “variance summary” in Output 2.13appears for the permeability data. Notice that 26.70% of the variability in the model effects(X-space) was summarized by the first PLS component and 43.36% was summarized bythe first two, etc. It may take as many as 20 components to adequately summarize thedescriptor space variability. The right-most columns have recorded the correlation betweenthe X-space score and the categorical response. For a discriminant application, this is notmeaningful since the group coding is arbitrary.

For purposes of this chapter, the X-space scores XSCR values are what we are primarilyinterested in (see Figure 2.10) since it is these new data that are then transferred to PROCDISCRIM for classification. These scores appear on the PROC PLS output as shown (fortwo components) below. The results of the classification, summarized in terms of eachgroup’s misclassification (“error”) rate, for various numbers of PLS components, aredisplayed in Table 2.6. These error rates are the so-called “apparent” misclassification ratesand are completely standard in discriminant analysis and can be read directly off the

Page 54: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 41

default SAS output. Both the error rates for a simple reclassification of the training set(called “re-substitution” rates on the SAS output) and the leave-one-out cross-validatedrates (called “cross-validated” rates on the SAS output) are reported. In all cases thepooled covariance matrix was used for the (linear) classification.

Table 2.6 Error Rates for PLS and PCA Followed by PROC DISCRIM on Component Scores (bolded entriesare the minimum (observed) misclassification rates)

Re-substitution method(Cross-validation method)

Total error rate 0-class error rate 1-class error rateNumber ofcomponents PLS PCA PLS PCA PLS PCA

2 0.3136 0.3390 0.2881 0.3220 0.3390 0.3559(0.3192) (0.3418) (0.2938) (0.3277) (0.3446) (0.3559)

10 0.2345 0.2966 0.2599 0.2768 0.2090 0.3164(0.2514) (0.3192) (0.2655) (0.2994) (0.2373) (0.3390)

20 0.2090 0.2885 0.2260 0.2768 0.1921 0.2881(0.2571) (0.3390) (0.2881) (0.2994) (0.2260) (0.3785)

30 0.2401 0.2345 0.2260 0.2429 0.2542 0.2260(0.3079) (0.2853) (0.3051) (0.2881) (0.3107) (0.2825)

40 0.2232 0.2345 0.2203 0.2712 0.2260 0.1977(0.3136) (0.3220) (0.3164) (0.3616) (0.3107) (0.2825)

50 0.2429 0.2458 0.2316 0.2599 0.2542 0.2316(0.3333) (0.3333) (0.3333) (0.3672) (0.3333) (0.3277)

PROC DISCRIM 0.2373 0.2316 0.2429with quasi-inverse (0.3192) (0.2938) (0.3446)

The following assessments are evident from Table 2.6:

• The cross-validated estimates of total misclassification rates suggest about a 25-30%rate. This rate is fairly consistent with the non-cross-validated direct re-substitutionmethod, although this latter method is overly optimistic, as expected.

• Practically stated, the permeable and non-permeable compounds are not well separated,even with quadratic boundaries, and the corresponding misclassification rates can beexpected to be fairly high in practice if one were to use the classification rule that wouldresult from this analysis.

• PROC DISCRIM with the quasi-inverse is recorded on the last line of the table. Noticethat the cross-validated misclassification rates for this alternative appear to do nobetter, perhaps worse, than PLS-LDA.

• As expected, PLS-LDA does a better job (with the exception of 30 components) thandoes PCA followed by an LDA. This is not really a surprise, of course, since the PLSdimension reduction step basically involved maximizing among-groups differences.

2.5.6 SummaryIn Section 2.5 we have briefly reviewed the formal sense in which an empirically obvioususe of PLS for the purposes of discrimination is actually optimal. This formal connection iseasily turned into a functional paradigm using PROC PLS. Direct connections to thetheory developed by Barker and Rayens (2003) can be had by invoking the SIMPLS optionin PROC PLS, but the connections would certainly hold (on an intuitive level) for otherrealizations of PLS as well.

Page 55: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

42 Pharmaceutical Statistics Using SAS: A Practical Guide

Again, what one does with the PLS scores after they are obtained seems to vary widelyby user. Some will want to then perform a formal discriminant analysis (as we did in theexample), while others will consider the problem “worked” at this point and simply do adhoc classification at the level of the scores, perhaps by identifying the sample group meanscore that is closest to an unknown observation.

The real point to the work of Barker and Rayens (2003) is that when a classicaldiscrimination is desired, but can’t be performed owing to singular covariances (eitherwithin or pooled), then initially reducing the dimension of the problem by identifyingoptimal linear combinations of the original features is a rational and well-acceptedapproach. In fact, it is common to use PCA to accomplish this first step. One of the pointsmade clear by Barker and Rayens (2003) is that PLS can always be expected to do a betterjob at this initial reduction than can PCA. It is an open question as to whether PLS usedin this fashion would outperform the SAS use of a quasi-inverse. One of the advantagesenjoyed by PLS-LDA (followed by PROC DISCRIM, perhaps) over using the quasi-inverseis that the sense in which these activities are optimal is well understood. It also would beinteresting to know if employing the quasi-inverse leads to understated non-cross-validatedmisclassification rates in general, or if that is just a characteristic that is somehow specificto this data set. That issue is a matter for future research, however, and will not bediscussed further here.

2.6 SummaryBoth boosting and partial least squares for linear discriminant analysis can be easilyimplemented using widely available SAS modules. The theoretical properties andperformance of each of these methods have been thoroughly researched by a number ofauthors. In this chapter, we have provided additional details about each method, haveprovided code to implement each method, and have illustrated their application on atypical drug discovery data set. Undoubtedly, as data structures become more complex,methods like these become extremely valuable tools for uncovering relationships betweendescriptors and response classification.

ReferencesBarker, M., Rayens, W.S. (2003). “A partial least squares paradigm for discrimination.” Journal of

Chemometrics. 17, 166–173.Bartlett, M.S. (1938). “Further aspects of the theory of multiple regression.” Proceedings of the

Cambridge Philosophical Society. 34, 33–40.Bauer, E., Kohavi, R. (1999). “An empirical comparison of voting classification algorithms:

Bagging, boosting, and variants.” Machine Learning. 36, 105–142.Breiman, L. (1996). “Bagging predictors.” Machine Learning. 26, 123–140.Breiman, L. (1998). “Arcing classifiers.” The Annals of Statistics. 26, 801–824.Breiman, L., Friedman, J., Olshen, R., Stone, C. (1984). Classification and Regression Trees.

Wadsworth.Frank, I., Friedman, J. (1993). “Statistical view of chemometric regression tools.” Technometrics.

35, 109–135.Freund, Y. (1995). “Boosting a weak learning algorithm by majority.” Information and

Computation. 121, 256–285.Freund, Y., Mansour, Y., Schapire, R.E. (2001). “Why averaging classifiers can protect against

overfitting.” Proceedings of the Eighth International workshop on Artificial Intelligence andStatistics. 1–8.

Freund, Y., Schapire, R.E. (1996a). “Game theory, on-line prediction and boosting.” Proceedings ofthe Ninth Annual Conference on Computational Learning Theory. 325–332.

Page 56: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 2 Modern Classification Methods for Drug Discovery 43

Freund, Y., Schapire, R.E. (1996b). “Experiments with a new boosting algorithm.” MachineLearning: Proceedings of the Thirteenth International Conference. 148–156.

Freund, Y., Schapire, R.E. (2000). “Discussion of the paper ‘Additive logistic regression: Astatistical view of boosting’ by Jerome Friedman, Trevor Hastie and Robert Tibshirani.” TheAnnals of Statistics. 38, 391–393.

Friedman, J. (1989). “Regularized discriminant analysis.” Journal of the American StatisticalAssociation. 405, 165–175.

Friedman, J., Hastie, T., Tibshirani, R. (2000). “Additive logistic regression: A statistical view ofboosting.” The Annals of Statistics. 38, 337–374.

Greene, T., Rayens, W.S. (1989). “Partially pooled covariance matrix estimation in discriminantanalysis.” Communications in Statistics. 18, 3679–3702.

Hastie, T. J., Buja, A., Tibshirani, R. (1995). “Penalized discriminant analysis.” Annals ofStatistics. 23, 73–102.

de Jong, S. (1993). “SIMPLS: An alternative approach to partial least squares regression.”Chemometrics and Intelligent Laboratory Systems. 18, 251–263.

Kansy, M., Senner, F., Gubernator, K. (1998). “Physicochemical high throughput screening:Parallel artificial membrane permeation assay in the description of passive absorptionprocesses.” J. Med. Chem. 41, 1007–1010.

Kearns, M., Valiant, L.G. (1989). “Cryptographic limitations on learning Boolean formulae andfinite automata.” Proceedings of the Twenty-First Annual ACM Symposium on Theory ofComputing. New York, NY: ACM Press.

Krieger, A., Long, C., Wyner, A. (2001). “Boosting noisy data.” Proceedings of the EighteenthInternational Conference on Machine Learning. 274–281.

Lavine, B.K., Davidson, C.E., Rayens, W.S. (2004). “Machine learning based pattern recognitionapplied to microarray data.” Combinatorial Chemistry and High Throughput Screening. 7,115–131.

Optiz, D., Maclin, R. (1999). “Popular ensemble methods: An empirical study.” Journal of ArtificialIntelligence Research. 11, 169–198.

Rayens, W.S., Greene, T. (1991). “Covariance pooling and stabilization for classification.”Computational Statistics and Data Analysis. 11, 17–42.

Rayens, W.S. (1990). “A role for covariance stabilization in the construction of the classical mixturesurface.” Journal of Chemometrics. 4, 159–170.

Ripley, B.D. (1996). Pattern Recognition and Neural Networks. Cambridge: Cambridge UniversityPress.

Schapire, R.E. (1990). “The strength of weak learnability.” Machine Learning. 5, 197–227.Schapire, R.E. (2002). “Advances in boosting.” Uncertainty in Artificial Intelligence: Proceedings of

the Eighteenth Conference. 446–452.Schapire, R.E., Freund, Y., Bartlett, P., Lee, W.S. (1998). “Boosting the margin: A new

explanation for the effectiveness of voting methods.” The Annals of Statistics. 26, 1651–1686.Tobias, R. (1997a). “An introduction to partial least squares regression.” TS-509, SAS Institute

Inc., Cary, NC. Available at http://www.sas.com/rnd/app/papers/pls.pdf.Tobias, R. (1997b). “Examples using the PLS procedure.” SAS Institute Inc., Cary, NC. Available

at http://www.sas.com/rnd/app/papers/plsex.pdf.Vapnik, V. (1996). The Nature of Statistical Learning Theory. New York: Springer-Verlag.Wold, H. (1966). Estimation of principal components and related models by iterative least squares.

Multivariate Analysis. New York: Academic Press.Wold, H. (1981). Soft modeling: The basic design and some extensions. Systems under indirect

observation, causality-structure-prediction. Amsterdam: North Holland.

Page 57: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

66

Page 58: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3

Model Building Techniques inDrug Discovery

Kimberly CriminThomas Vidmar

3.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45

3.2 Example: Solubility Data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46

3.3 Training and Test Set Selection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47

3.4 Variable Selection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51

3.5 Statistical Procedures for Model Building . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58

3.6 Determining When a New Observation Is Not in a Training Set . . . . . . . . . . . . . . . . . . . . . 61

3.7 Using SAS Enterprise Miner . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63

3.8 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67

This chapter focuses on techniques used in the early stages of drug discovery to optimizethe model building process. The main topics discussed in this chapter are selecting trainingand test sets, selecting variables, and determining when the prediction of a new observationis valid. Statistical procedures that are used to build models are also reviewed. For each ofthe main topics we provide background, discuss a new procedure, and provide guidance onimplementation in SAS. We apply these methods to a real drug discovery data set.

3.1 IntroductionComputational models are used in many stages of drug discovery to predict structureactivity and property relationships. The models built are based on the principle thatcompounds with similar structures are expected to have similar biological activities(Golbraikh and Tropsha, 2002). In the early stage of drug discovery, high throughputscreens are used to identify a compound’s activity against a specific target. Computationalmodels used at this stage need to predict a compound’s activity but the model does notneed to be interpretable. Once a series of compounds has been identified as being activeagainst the biological target, then the goal is to optimize the compound in terms ofstructure properties such as solubility, permeability, etc. At this stage, it is useful to have acomputational model that can be interpreted by the scientists so they understand theimpact on the property when a particular feature of the structure is changed. For example,suppose solubility has an inverse relationship with a certain feature of the structure, such

Kimberly Crimin is Senior Principal Biostatistician II, Early Development, Wyeth, USA. Thomas Vidmar is Senior Director,Midwest Nonclinical Statistics, Pfizer, USA.

Page 59: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

46 Pharmaceutical Statistics Using SAS: A Practical Guide

as number of rotatable bonds; if the goal is to increase solubility then the team knows thatdecreasing the number of rotatable bonds will increase solubility. In this chapter we focuson methods the statistician can use to optimize the model building process. As George Box(Box et al., 1978) once said “All models are wrong, but some are useful”.

To build a model, a statistician will perform the following steps: divide the data intotraining and test sets, select the variables, and select the best statistical tool for prediction.There exists a vast body of literature on statistical procedures for model building, a few ofwhich we discuss here. In this chapter, we focus on statistical techniques that can optimizethe model building process independent of the statistical procedure used to build the model.

A computational model is evaluated on the ability to predict future observations. Onemeasure of this performance is the prediction error of the model calculated on anindependent test set; therefore, it is important that the test set be representative of thetraining set. Otherwise, the prediction error obtained from the test set will not beindicative of the model’s performance.

Molecular descriptors describe geometric, topological, and electronic properties of thecompound. The molecular descriptors used to build structure activity and propertyrelationship models are often computer generated and many programs are available togenerate molecular descriptors. Because of this, the modeler is often faced with hundreds, ifnot thousands, of descriptors. Since the molecular descriptors are based on attributes of thestructure many of these descriptors are highly correlated. To build a model that isinterpretable, the goal of variable selection is to choose descriptors that are highlypredictive of the response and independent of one another. A computational model builtwith independent descriptors will be more interpretable than a model built with correlateddescriptors.

When using a model to predict a response for a new set of data, it is important to knowhow different the new data are from the data used to train the model. If the new set ofdata is “far” from the data used to train and test the model, then one might considerretraining the model because the error in the prediction will be large.

In Section 3.2 we provide an example using a real drug discovery data set. In Section 3.3we review methods for training and test set selection, and we discuss a novel technique thatwe found useful. In Section 3.4 we provide an overview of variable selection techniques andprovide a new technique. In Section 3.5 we briefly review statistical procedures for modelbuilding. In Section 3.6 we discuss a simple procedure that can be used to determine if anew observation is in the descriptor space the model was trained on. And in Section 3.7 weuse SAS Enterprise Miner to build a computational model.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

3.2 Example: Solubility DataSolubility is an important physical-chemical property in drug discovery. In the simplestdefinition, the solubility of a solute is the maximum amount of the solute that can dissolvein a certain amount of solvent. Intrinsic solubility is the solubility of the neutral form ofthe salt; the salt has not separated into ions. Some of the factors affecting solubility are:temperature, molecular weight, number of hydrogen bonds, and polarity.

For a solvent to dissolve a solute, particles of the solvent must be able to separate theparticles of the solute and occupy the intervening spaces. Water is a polar substance andpolar solvents can generally dissolve solutes that are ionic. Dissolving takes place when thesolvent is able to pull ions out of their crystal structure. Separation of ions by the action ofa solvent is called dissociation; ions are dissociated by the water molecules and spreadevenly throughout the solution.

Page 60: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 47

Estimating solubility then becomes a bit more complicated when compounds have anionized group because multiple forms of the compound exist with varying solubilities. Theterm aqueous solubility refers to the solubility of all forms of a compound. It is the sum ofthe solubility for the neutral compound plus the solubility of each ionized form of thecompound; aqueous solubility is, therefore, a function of pH. Since most drugs are eitherweakly acidic or basic, it is important to use the intrinsic solubility when building acomputational model to predict solubility.

The example that we use in this chapter is a solubility data set (SOLUBILITY data setcan be found on the book’s companion Web site). The solubilities were estimated using theold-fashioned method. An excessive amount of the compound was added to a flask ofwater; the flask was shaken and the amount of remaining compound measured. Theamount of the compound dissolved is the solubility of that compound. In this data set,there are 171 compounds. To limit the complexity of the example, only one softwarepackage was used to generate the molecular descriptors. The goal is to build aninterpretable computational model that predicts the solubility of new compounds.

3.3 Training and Test Set SelectionIn order to build reliable and predictive models, care must be taken in selecting trainingand test sets. When the test set is representative of the training set, one can obtain anaccurate estimate of the model’s performance. Ideally, if there is sufficient data, themodeler can divide the data into three different data sets: training set, validation set, andtest set. The training set is used to train different models. Then each of these models isapplied to the validation set and the model with the minimum error is selected as the finalmodel and applied to the test data set to estimate the prediction error of the model. Moreoften than not, there is insufficient data to apply this technique.

Another method used to calculate the model prediction error is cross-validation. In thismethod, the data are divided into K parts, each approximately the same size. For each ofthe K parts, the model is trained on the remaining K − 1 parts and then applied to the Kthpart and the prediction error calculated. The final estimate of the model prediction error isthe average prediction error over all K parts. The biggest question associated with thismethod is how to choose K. If K is chosen to be equal to the sample size, then the estimateof error will be unbiased, but the variance will be high. As K decreases, the variancedecreases but the bias increases, so one needs to find the appropriate trade-off between biasand variance. See Hastie, Tibshirani, and Friedman (2001) for more information.

Another method which is similar to cross-validation is the bootstrap method. Thismethod takes a bootstrap sample from the original data set and uses the data to train themodel. Then, to calculate the model prediction error, the model is applied to either theoriginal data set or to the data not included in the bootstrap sample. This procedure isrepeated a large number of times and the final prediction error is the average predictionerror over all bootstrap samples. The main issue with this method is whether the originaldata set is used to calculate the prediction error or only the observations not included inthe bootstrap sample. When the original sample is used, the model will give relatively goodpredictions because the bootstrap sample (training set) is very similar to the originalsample (test set). One solution to reduce this bias is to use the “0.632 estimator”. For moreinformation on this method see Efron and Tibshirani (1998).

3.3.1 Proposed Procedure for Selecting Training and Test SetsThe methods discussed above are easy to implement and will give adequate estimates ofthe prediction error provided there are no outlying points in either the response space orthe descriptor space. Often in drug discovery data sets there are points of high influence oroutliers, so excluding them from the training set is an important consideration. Therefore,

Page 61: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

48 Pharmaceutical Statistics Using SAS: A Practical Guide

we developed a training and test set selection procedure that limits the number of outliersand points of influence in the training set. In this section we provide an overview of theproposed method and apply it to the solubility data set.

Our goal is to develop a training and test set selection algorithm that accomplishes thefollowing goals:

• Select a test set that is representative of the training set.• Minimize the number of outliers and influential points in the training set.

Suppose a data set contains only one descriptor. Then it would be fairly easy to comeup with an algorithm that meets the above goals. For instance, the modeler could use atwo-dimensional plot to identify outliers and points of influence, assign these to the testset, and then take a random sample of the remaining points and assign them to thetraining set. To view the data set selection, the modeler could again use a two-dimensionalplot and if the test set did not visually appear to be representative of the training set, themodeler could take another random sample.

The above procedure is simple and easy to explain to scientists and accomplishes thetwo goals when p = 1, but it is not easily extendable for p > 1 because the modeler wouldhave to view (p+1

2 ) two-dimensional projections to identify outliers and points of influence.Furthermore, selecting a representative sample in p + 1-dimensional space would beimpossible unless the following were true: instead of using each descriptor, a summarymeasure for each observation in the descriptor space were used, for instance, the leveragevalue of each observation. Recall that the leverage value for the ith observation is thedistance between the observation and the center of the descriptor space. By using theleverage values and the responses, we have reduced the space down to 2 from p + 1 and thealgorithm discussed above (when p = 1) could be used to develop an algorithm for trainingand test set selection.

The basic algorithm for the proposed procedure is defined as follows:

1. Calculate the leverage value for each observation.2. Bin the leverage values and the responses and then combine the bins into cells. Two

possible ways to calculate the bin size are discussed below.3. Potential outliers and points of influence are identified as points outside a (1 − α)%

confidence region around the responses and the leverage values. These points areassigned to the test set.

4. For each cell, take a random selection of the points within the cell and assign them tothe training set; assign the other points to the test set.

To enhance the above algorithm, the user can select how the bin size is calculated. Oneoption, referred to as the normal bin width, defines the bin width to be:

W = 3.49σN−1/3,

where N is the number of samples and σ is the standard deviation of either the responses orthe leverages. The second option, referred to as robust bin width, defines the bin width to be:

W = 2(IQR)N−1/3,

where IQR is the interquartile range of either the response or the leverage values and N isdefined above. We have found that the robust bin width results in more cells being defined,and we suggest the normal bin width be used if the data are sparse. For discussions on theoptimal bin size see Scott (1979).

Page 62: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 49

After running the basic algorithm on a number of examples, we added two additionalfeatures to improve it. The user has an option of pre-allocating particular observations toeither the training or test data sets. The other feature we added allows the user to specifythe seed used to initialize the random number generator. This allows the user to get thesame results on multiple runs.

3.3.2 TRAINTEST ModuleThe algorithm described above was implemented in a SAS/IML module (TRAINTESTmodule) that can be found on the book’s companion Web site. There are seven inputs tothis module:

• the matrix of descriptors• the vector of responses• the type of bins to use (normal or robust)• the desired proportion of observations in the training set• a vector indicating the data set each observation should be in (if the user doesn’t want to

pre-allocate any observations, then this vector should be initialized to a vector of zeros)• the level of the confidence region around the leverage values and the response• the seed

(If the seed is set to zero, then the module allows SAS to initialize the random numbergenerator; SAS uses the system time for initialization.)

The TRAINTEST module produces three outputs plus a visual display of the trainingand test set selections. The three outputs are: a vector indicating the data set eachobservation belongs to, the vector of leverage values, and the observed proportion ofobservations in the training set. Finally, the module creates a SAS data set (PLOTINFO)that can be used to create a plot of leverage values versus responses. The PLOTTYPEvariable in this data set serves as a label for individual observations: PLOTTYPE=1 if theobservation is in the training set, PLOTTYPE=2 if the observation is in the test set, andPLOTTYPE=3 if the observation is an outlier.

3.3.3 Analysis of Solubility DataThe SOLUBILITY data set that can be found on the book’s companion Web site has 171observations (compounds) and 21 possible descriptors. The response variable, the log of themeasured solubility of the compound, is continuous. The objective is to divide the data setinto two data sets, one for training the model and the other for testing the model. Basedon the size of the data set, we wanted approximately 75% of the observations to beallocated to the training set and we considered any point outside the 95% confidence regionto be an outlier. Program 3.1 contains the SAS code to run the training and test setselection module on the solubility data.

Program 3.1 first reads in the SOLUBILITY data set and stores the descriptors into amatrix X and the response into a vector y. After that, it calls the TRAINTEST moduleand, based on the allocations returned from the module, the full data set is divided intothe training and test sets.

Program 3.1 Training and test set selection in the solubility example

proc iml;* Read in the solubility data;use solubility;read all var (’x1’:’x21’) into x;read all var {y} into y;

Page 63: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

50 Pharmaceutical Statistics Using SAS: A Practical Guide

* Initialize the input variables;type=1;prop=0.75;level=0.95;init=j(nrow(x),1,0);seed=7692;* Next two lines of code are not executed on the first run,;* only the second run to allocate certain observations to the training set;*outindi={109, 128, 139, 165};*init[outindi,1]=1;call traintest(ds,hi,obsprop,x,y,type,prop,init,level,seed);print obsprop[format=5.3];* Using data set definitions returned in DS, create two data sets;trindi=loc(choose(ds=1,1,0))‘;tsindi=loc(choose(ds=2,1,0))‘;trx=x[trindi,]; try=y[trindi,1]; trxy=trx||try;tsx=x[tsindi,]; tsy=y[tsindi,1]; tsxy=tsx||tsy;* Number of observations in the train and test data set;ntrain=nrow(trxy); ntest=nrow(tsxy);print ntrain ntest;create soltrain from trxy[colname=(’x1’:’x21’||’y’)];append from trxy;close soltrain;create soltest from tsxy[colname=(’x1’:’x21’||’y’)];append from tsxy;close soltest;quit;

* Vertical axis;axis1 minor=none label=(angle=90 "Response") order=(-7 to 5 by 2) width=1;* Horizontal axis;axis2 minor=none label=("Leverage") order=(0 to 0.8 by 0.2) width=1;symbol1 i=none value=circle color=black height=5;symbol2 i=none value=star color=black height=5;symbol3 i=none value=dot color=black height=5;proc gplot data=plotinfo;

plot response*leverage=plottype/vaxis=axis1 haxis=axis2 frame nolegend;run;quit;

Output from first run of Program 3.1

OUTINDI

14 96 109 128 139 140 151 155 165

OBSPROP

0.688

NTRAIN NTEST

117 53

Output 3.1 lists the IDs of the outlying observations (OUTINDI), observed proportion(OBSPROP) and number of observations in the training and test sets (NTRAIN andNTEST) after the first run of Program 3.1. Figure 3.1 contains the plot of the pointsassigned to each data set.

Page 64: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 51

Figure 3.1 Initial assignment of observations, training set (circle), test set (star) and outlier (dot)

Res

pons

e

-7

-5

-3

-1

1

3

5

Leverage

0.0 0.2 0.4 0.6 0.8

Output from second run of Program 3.1

OUTINDI

14 96 140 151 155

OBSPROP

0.712

NTRAIN NTEST

121 49

The first run of the program allocated 117 observations (68.8%) to the training set and53 observations to the test set. The proportion of the observations assigned to the trainingset was a bit lower than the desired proportion. Upon visual inspection of the data setdefinitions, Figure 3.1, it was decided to run the program again only this time allocating 4observations identified as outliers to the training set (observations 109, 128, 139 and 165).Figure 3.2 contains the plot after the second run of Program 3.1 and the output shows theIDs of the outlying observations, observed proportion, and number of observations in eachdata set after the second run. After the second run, 121 observations (71.2%) were assignedto the training set and 49 observations to the test set.

3.4 Variable SelectionThe data used to develop computational models can often be characterized by a fewobservations and a large number of measured variables, some of which are highlycorrelated. Traditional approaches to modeling these data are principle componentregression (PCR) and partial least squares (PLS) (Frank and Friedman, 1993). The factorsobtained from PCR and PLS are usually not interpretable, so if the goal is to develop aninterpretable model, these are not as useful. By considering the loadings given to each

Page 65: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

52 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 3.2 Final assignment of observations, training set (circle), test set (star) and outlier (dot)

Res

pons

e

-7

-5

-3

-1

1

3

5

Leverage

0.0 0.2 0.4 0.6 0.8

variable, these methods can be used to prune the variables, although the criteria forremoving variables can be inconsistent between data sets.

Other popular variable selection procedures are tied to model selection and are oftenused when fitting multiple linear regression models, although they can be used with othermodels, such as a linear discriminant model. A few of the more popular procedures arediscussed below. Shrinkage methods are also available to the modeler. Instead of selectingvariables, shrinkage methods shrink the regression coefficients by minimizing a penalizederror sum of squares, subject to a constraint. In ridge regression, a common shrinkagemethod, the L2 norm is used as the constraint, and Lasso, another shrinkage method, theL1 norm is used as the constraint.

Best K models examine all possible models and the top K models are selected forfurther investigation by the modeler. The top models are selected using a measure of fitsuch as adjusted R2 (R2

a) or Mallow’s Cp. Since all possible models are examined, thesample size, n, must be strictly greater than the number of descriptors, p. Examining allpossible models is computationally intensive and, realistically, this method should be usedonly if p is small. A rule of thumb is p < 11.

Best subset algorithms, such as leaps and bounds (Furnival and Wilson, 1974), do notrequire the examination of all possible subsets, but use an efficient algorithm to search forthe K best models using a statistical criteria such as R2

a or Mallow’s Cp. It is recommendedthat other variable selection procedures be used if p > 60.

Stepwise procedures, including forward selection and backward elimination, developmodels at each step by adding (forward selection) or deleting (backward elimination) thevariable that has the largest (forward selection) or smallest (backward elimination) F-valuemeeting a particular pre-determined limit. Stepwise procedures are commonly used to buildcomputational models.

The variable selection methods mentioned above are dependent on the type of modelbeing fit and often select variables for the final model which are highly correlated. This isnot an issue unless the modeler is interested in an interpretable model. In the followingsection we present a variable selection method that is based on a stable numerical linearalgebra technique. Our technique ranks the predictor variables in terms of importance,taking into account the correlation with the response and the predictor variables previouslyselected. Our technique deals with the collinearity problem by separating correlated

Page 66: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 53

variables during the ranking. Our procedure is a modification of the variable selectionprocedure proposed by Golub and Van Loan (1996) and is not dependent on the modelerpre-specifying the type of model.

3.4.1 Variable Selection AlgorithmThe proposed variable selection algorithm is defined below:

1. Center the matrix of descriptors, X.2. Center the vector of responses, Y.3. Calculate Y, where each column is the projection of y onto xi, where x′

i is the ith row ofX.

4. Use the Singular-Value Decomposition to calculate the rank of Y.5. Apply the QR decomposition with column-pivoting to Y.

The output from the QR decomposition algorithm when column pivoting is invoked is anordering of the columns of Y. The number of descriptors to use in the model can beobtained from the magnitude of the singular values of the ordered descriptor matrix. Thisis similar to using a scree plot in PCR to determine the number of directions to use.

The QR decomposition of the matrix X is X = QR, where Q is an orthogonal matrixand R is upper triangular. The matrix Q if formed from the product of p Householdermatrices, Hi, i = 1, . . . , p. A Householder matrix is an elementary reflector matrix. Whencolumn-pivoting is incorporated into the QR algorithm, prior to calculating eachHouseholder matrix, the column with the largest norm is moved to the front of the matrix.Since the matrix on which the QR algorithm operates is Y, at each stage the columnmoved to the front of the matrix will correspond to the descriptor with the highestcorrelation with y but relatively independent of the previously selected descriptors, xi. Inother words, the first column selected will be the descriptor that has the highest correlationwith the response. The second column selected will be the descriptor with the highestcorrelation with the response and the lowest correlation with the first descriptor selected.This continues until all the descriptors have been ordered.

3.4.2 ORDERVARS ModuleThe variable ordering procedure described in Section 3.4.1 was implemented in a SAS/IMLmodule (ORDERVARS module) that can be found on the book’s companion Web site. Theinputs to this module are the matrix X of descriptors and the vector y of responses. Theoutputs are the variable ordering, the rank of X, and a scree plot of the singular values.

The ORDERVARS module starts by centering X and y and then calculates the matrixY. The QR-algorithm implemented in SAS/IML (QR subroutine) is called.Column-pivoting is invoked in the QR subroutine by initializing the input vector ORD toall zeros. The ORDERVARS module also creates a SAS data set (SINGVALS) that storesthe computed singular values.

3.4.3 Example: Normally Distributed ResponsesAs an illustrative example, suppose x is distributed N5(0,Σ), where

Σ =

⎡⎢⎢⎢⎢⎣1 0.75 0.05 0 0

0.75 1 0 0 00.05 0 1 0.98 00 0 0.98 1 00 0 0 0 1

⎤⎥⎥⎥⎥⎦ .

Page 67: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

54 Pharmaceutical Statistics Using SAS: A Practical Guide

In this example, x1 is correlated with x2 and x3 is highly correlated with x4. Take 25samples from the above distribution and define

y = 2x1 + 1.5x3 + x5 + ε,

where ε is distributed N(0, 1). The response is a linear function of three of the fivedescriptors, namely, x1, x3, x5. A variable selection procedure should select 1, 3, 5 as thefirst three variables.

Program 3.2 provides the code to randomly generate the descriptors for this exampleand calls the ORDERVARS module on the simulated data. The program starts byinitializing the covariance matrix, randomly generating the matrix of descriptors, and thencalculating y. To randomly generate data from a multivariate distribution with mean μ andpositive-definite covariance matrix Σ, the following transformation was used

x = Σ1/2z + μ,

where z is a p × 1 vector of observations, zi distributed N(0, 1), and the matrix Σ1/2 is thesquare root matrix of Σ. The square root matrix of a positive definite matrix can becalculated from the spectral decomposition of Σ as:

Σ1/2 = CD1/2C ′,

where C is the matrix of eigenvectors and D is a diagonal matrix whose elements are thesquare-root of the eigenvalues.

Program 3.2 Variable ordering in the simulated example

proc iml;* Initialize Sigma;call randseed(564);sig={1 0.75 0.05 0 0,

0.75 1 0 0 0,0.05 0 1 0.98 0,0 0 0.98 1 0,0 0 0 0 1};

* Calculate sigma^{1/2};call eigen(evals,evecs,sig);sig12=evecs*diag(evals##0.5)*evecs‘;* Generate the matrix of descriptors, x;n=25;x=j(n,5,0);do i=1 to n by 1;d=j(1,5,.);call randgen(d,’normal’,0,1);x[i,]=(sig12*d‘)‘;end;err=j(1,n,.);* Randomly generate errors and calculate y;call randgen(err,’normal’,0,1);y=j(n,1,0);y=2*x[,1] + 1.5*x[,3] + x[,5] + err‘;* Concatenate x and y into one matrix;xy=x||y;* Calculate the correlation matrix;cormat=j(6,6,.);cormat=corr(xy);print cormat[format=5.3];

Page 68: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 55

* Save example data set;create test from xy[colname={’x1’ ’x2’ ’x3’ ’x4’ ’x5’ ’y’}];append from xy;close test;use test;read all var (’x1’:’x5’) into x;read all var {y} into y;* Call the variable selection procedure;call ordervars(x,y,order,rank);* Print rank and variable ordering *;print rank;print order;quit;

Output from Program 3.2

CORMAT

1.000 0.808 0.129 0.044 0.175 0.7460.808 1.000 0.014 -.037 0.062 0.5110.129 0.014 1.000 0.977 0.294 0.6030.044 -.037 0.977 1.000 0.308 0.5500.175 0.062 0.294 0.308 1.000 0.5760.746 0.511 0.603 0.550 0.576 1.000

RANK

5

ORDER

1 3 5 2 4

After randomly generating the data, the correlation matrix is calculated for the matrix[Xy]. The correlation matrix is given in the Output 3.2. The last row of the correlationmatrix corresponds to y. Looking at the correlations, we see that x1 has the highestcorrelation with the response y and should be selected first by the variable orderingprocedure. Output 3.2 also contains the variable ordering produced by the ORDERVARSmodule. The ordering of the variables is: x1, x3, x5, x2, and x4.

3.4.4 Forward SelectionLet’s compare the above results to the results obtained from forward selection. Program 3.3contains the code to run forward selection in SAS using the REG procedure. The TESTdata set used in this program was created in Program 3.2.

Program 3.3 Forward selection on simulated data

proc reg data=test;model y = x1 x2 x3 x4 x5/selection=forward;run;

Page 69: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

56 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 3.3

All variables have been entered into the model.

Summary of Forward Selection

Variable Number Partial ModelStep Entered Vars In R-Square R-Square C(p) F Value Pr > F

1 x1 1 0.5572 0.5572 93.1599 28.94 <.00012 x4 2 0.2678 0.8250 26.1153 33.67 <.00013 x5 3 0.0956 0.9206 3.4650 25.29 <.00014 x2 4 0.0036 0.9242 4.5439 0.94 0.34325 x3 5 0.0021 0.9263 6.0000 0.54 0.4698

Output 3.3 contains the output from Program 3.3. Using forward selection, the variablesare entered x1, x4, x5, x2, x3, which is different from the variables used to generate y.

3.4.5 Variable Ordering in the Solubility DataProgram 3.4 executes the ORDERVARS module on the solubility training set. Thesolubility training and test sets (SOLTRAIN and SOLTEST data sets) were generated inProgram 3.1. At the end of Program 3.4, the training and test data sets are created usingthe new variable ordering, and they are stored as SAS data sets (SOLTRUSE andSOLTSUSE data sets).

Program 3.4 Variable ordering in the solubility data

proc iml;use soltrain;read all var (’x1’:’x21’) into x;read all var {y} into y;* Call the variable ordering module;call ordervars(x,y,order,rank);print rank;print order;* Store the variable ordering;create solorder var{order};append;close solorder;* Create the training and test sets using the new variable order;* Store the training and test sets;trxy=x[,order[1,]]||y;create soltruse from trxy[colname=(’x1’:’x21’||’y’)];append from trxy;close soltruse;use soltest;read all var (’x1’:’x21’) into tsx;read all var {y} into tsy;tsxy=tsx[,order[1,]]||tsy;create soltsuse from tsxy[colname=(’x1’:’x21’||’y’)];append from tsxy;close soltsuse;* Number of observations in the train and test data set;ntrain=nrow(trxy); ntest=nrow(tsxy);print ntrain ntest;quit;

Page 70: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 57

* Vertical axis;axis1 minor=none label=(angle=90 "Singular value") order=(0 to 12000 by 2000) width=1;* Horizontal axis;axis2 minor=none label=("Index") order=(1 to 21 by 1) width=1;symbol1 i=none value=dot color=black height=5;proc gplot data=singvals;

plot singularvalues*index/vaxis=axis1 haxis=axis2 frame;run;quit;

Figure 3.3 Singular values in the solubility data set

Sin

gula

r va

lue

0

2000

4000

6000

8000

10000

12000

Index

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

Output from Program 3.4

RANK

21

ORDERCOL1 COL2 COL3 COL4 COL5 COL6 COL7

ROW1 8 13 3 1 21 9 19

ORDERCOL8 COL9 COL10 COL11 COL12 COL13 COL14

ROW1 12 20 2 4 5 16 10

ORDERCOL15 COL16 COL17 COL18 COL19 COL20 COL21

ROW1 6 17 18 7 14 15 11

NTRAIN NTEST

121 49

Page 71: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

58 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 3.3 is a plot of the singular values for each descriptor. Based on this plot itappears only five descriptors are needed to build a predictive model, but sequentialregression should probably be used to determine the number of descriptors needed in themodel. In sequential regression the variables can be added in order, provided that a certainstatistical criterion is met. An example is provided in Section 3.5.1. Output 3.4 containsthe variable ordering (ORDER) as well as the number of observations in the training andtest data sets (NTRAIN and NTEST).

3.5 Statistical Procedures for Model BuildingIn this section we review a number of procedures that can be used to build statisticalmodels. The potential model builder will need to obtain a substantial toolbox since no onestatistical tool will work on every set of data. As a corollary to the previous statement, thenature of data will help determine the appropriate statistical model-building tool. See TwoCrows Corporation (1999) for a review of model building procedures used to extractpredictive patterns in data.

3.5.1 Multiple Linear RegressionMultiple linear regression has certainly been the workhorse for building predictive modelsfor many years. It is appropriate for the situation where there are many more observationsthan descriptors and where the dependent variable is continuous. Models take the form of:

y = β0 + β1x1 + β2x2 + · · · + βpxp + e,

where y is the dependent variable, x1, . . . , xp are the independent variables, and e is therandom error. If a linear combination of the descriptors can approximate the response, thenthis formulation works well and is simple to perform. Terms such as xn

i or xixj are alsoallowed. PROC REG is appropriate and allows for variations of this simple procedure, suchas when terms are added or deleted in a stepwise fashion. Program 3.3 illustrated the useof PROC REG with forward selection.

PROC REG can also be used interactively. After you specify a model in PROC REGwith a RUN command, but not a QUIT command, you can use a variety of othercommands interactively such as ADD, DELETE, PRINT. See the PROC REGdocumentation for more information. To demonstrate the utility of this, Program 3.5provides the commands for running PROC REG interactively on the solubility training set.In Program 3.5, the initial model fit contains the five most important variables, x1, . . . , x5(they are included in the MODEL statement) and the VAR statement specifies the othervariables that might be added to the model. The following commands add variables to theinitial model with the ADD statement. The QUIT command ends PROC REG. The resultsfor the final model are included in Output 3.5. The output contains, for the intercept andeach of the 11 variables added to the model interactively, the parameter estimate, thestandard error of the estimate, and the one-degree of freedom test.

Page 72: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 59

Program 3.5 Using PROC REG interactively on the solubility data

proc reg data=soltruse;model y = x1 x2 x3 x4 x5;var x6 x7 x8 x9 x10 x11 x12 x13 x14;run;add x6 x7;print;run;add x8 x9;print;run;add x10 x11;print;run;quit;

Output from Program 3.5

Parameter Estimates

Parameter StandardVariable DF Estimate Error t Value Pr > |t|

Intercept 1 -3.40278 3.57476 -0.95 0.3433x1 1 -0.00771 0.00198 -3.89 0.0002x2 1 -0.00851 0.00162 -5.25 <.0001x3 1 0.77612 0.22350 3.47 0.0007x4 1 0.08929 0.04636 1.93 0.0567x5 1 0.23927 0.11684 2.05 0.0430x6 1 0.07884 0.03048 2.59 0.0110x7 1 7.36932 3.37147 2.19 0.0310x8 1 -0.00510 0.00126 -4.06 <.0001x9 1 -0.40608 0.12531 -3.24 0.0016x10 1 0.63875 0.29842 2.14 0.0345x11 1 0.88649 0.20612 4.30 <.0001

3.5.2 Logistic RegressionLogistic regression is a generalization of linear regression in that it is formulated to use abinary (0 or 1) dependent variable. It uses the log odds or logit transformation:

log(

Pi

1 − Pi

)= β0 + β1x1 + β2x2 + · · · + βpxp + e,

where Pi is the probability of the event occurring. The model above assumes the log oddsratio is a linear function of the predictors. It is easy to see that similar pros and cons existbetween logistic regression and multiple linear regression discussed earlier.

There are several SAS procedures that you can use to perform logistic regression such asthe LOGISTIC, CATMOD, and GENMOD procedures.

3.5.3 Discriminant AnalysisDiscriminant analysis is an extremely old statistical technique developed by R. A. Fisher inthe 1930’s and used to classify the famous Iris data set. This procedure determines thehyper-planes that separate or discriminate between the various classes in the data. It is avery simple technique to use and interpret since an observation is on one side of the plane

Page 73: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

60 Pharmaceutical Statistics Using SAS: A Practical Guide

or the other. However, there are assumptions such as normality of the descriptors, whichcan be problematic, and the fact that the boundaries separating the different classes arelinear may reduce its discrimination ability.

There are several SAS procedures that you can use to perform discriminant analysissuch as the DISCRIM, CANDISC, and STEPDISC procedures.

3.5.4 Generalized Additive RegressionThe standard linear regression model is used quite often for building models. It isextremely easy to use but suffers from the fact that, in real life, many effects are not linear.The generalized additive regression model offers a flexible statistical technique that canaccommodate factors that are not linear. Additionally, generalized additive regression is aninteresting tool because it allows both continuous and discrete responses.

Recall that the standard linear regression model assumes the expected value of y has thelinear form:

E(y|X) = f(x1, . . . , xp) = β0 + β1x1 + · · · + βpxp.

The additive model generalizes the linear model by modeling the expected value of y as

E(y|X) = s0 + s1(x1) + s2(x2) + · · · + sp(xp),

where si, i = 1, . . . , p are smooth functions. These function are estimated in anonparametric fashion. Generalized additive models are able to fit both discrete andcontinuous responses by allowing for a link between f(x1, . . . , xp) and the expected value ofy. Additional details may be found in the GAM procedure documentation.

This procedure has a lot of appeal and can be very useful. Since it is new to SAS, we willillustrate its use on the solubility training data set. Program 3.6 contains the SAS code toread the solubility training set and fit the model using five variables. The GAM proceduredetermines the values of smoothing parameters for each of the variables. For the solubilityexample, the SPLINE smoothing effect was chosen for each variable. The other optionsavailable are LOESS, SPLINE2, or PARAM. The LOESS option fits a local regression withthe variable, the SPLINE2 option fits a bivariate thin-plate spline to two variables, andPARAM specifies a parametric variable; a smoothing function is not applied to the variable.

Program 3.6 Analysis of the solubility data using PROC GAM

proc gam data=soltruse;model y=spline(x1) spline(x2) spline(x3) spline(x4) spline(x5)/dist=normal;run;quit;

Page 74: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 61

Output from Program 3.6

Regression Model AnalysisParameter Estimates

Parameter StandardParameter Estimate Error t Value Pr > |t|

Intercept -0.73649 0.36152 -2.04 0.0442Linear(x1) -0.00741 0.00118 -6.31 <.0001Linear(x2) -0.00677 0.00158 -4.30 <.0001Linear(x3) 0.79513 0.19670 4.04 0.0001Linear(x4) 0.01799 0.03777 0.48 0.6349Linear(x5) 0.17723 0.10212 1.74 0.0857

Smoothing Model AnalysisAnalysis of Deviance

Sum ofSource DF Squares Chi-Square Pr > ChiSq

Spline(x1) 3.00000 14.958834 23.9431 <.0001Spline(x2) 3.00000 5.345832 8.5565 0.0358Spline(x3) 1.00000 0.038719 0.0620 0.8034Spline(x4) 3.00000 1.252530 2.0048 0.5714Spline(x5) 3.00000 7.541784 12.0714 0.0071

Output 3.6 provides the output from Program 3.6. From the output we can see that x1,x2, and x3 have significant linear trends; p-values for the corresponding linear tests aresignificant at 5%. From the analysis of deviance table, the chi-square tests are significantfor x1, x2, and x5, indicating that these variables need to be smoothed.

A plot of the smoothing components with 95% confidence bands for each of the fivedescriptors can be created using the ODS GRAPHICS statement as shown below:

ods html;ods graphics on;proc gam data=soltruse plots(clm commonaxes);

model y=spline(x1) spline(x2) spline(x3) spline(x4) spline(x5)/dist=normal;run;quit;

ods graphics off;ods html close;

The plot generated using ODS GRAPHICS is not provided because it is notpublication-quality and, due to the experimental nature of PROC GAM, the option to savethe smoothed components to a SAS data set is not yet available.

3.6 Determining When a New Observation Is Not in a Training SetOften in drug discovery, a computational model is built for a non-statistician, and the userrelies on easily understood diagnostics and graphical representations to assess the accuracyof the prediction.

Once a model is in use, a common question asked of the statistician is, “How accurate isthe prediction?” If the model is providing the user with a quantitative estimates, then theerror associated with the prediction can be used to answer this question. This measure

Page 75: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

62 Pharmaceutical Statistics Using SAS: A Practical Guide

takes into account the distance between the new observation and the training set as well asthe error in the model. If the model is providing the user with a categorical response and alinear discriminant model is used, then a bar chart of the probability that an observation isin each group can be provided to the user.

Another measure that can be provided to the user is the leverage value. Recall theleverage value is simply the distance between the new observation and the centroid of thetraining data set. If the leverage value of a new observation is greater than the maximumobserved leverage value in the training set, then the prediction for the new observation willbe unreliable.

Also, it is important for the modeler to keep track of the data sets the model is beingapplied to. If the data is far from the data the model was trained on, then the modelershould think about retraining the data sets so the predictions will be reliable.

Once the statistician has provided the model, along with easily understood diagnostics,to the team, the statistician should also monitor how the model is being used. If thechemistry space the model is being used on is different from the chemistry space the modelwas trained on, then the statistician should consider retraining the model to provide theteam with a more useful model. This monitor can easily be done by keeping track of theleverage value from each prediction and by using a flag to indicate whether the predictionwas an extrapolation or not.

As an example, let’s determine if the observations in the solubility test set are includedin the solubility training space. Program 3.7 contains the code to fit the selected model tothe test set and store the residuals in a SAS data set (RESDS data set). This isaccomplished using PROC REG.

Program 3.7 also contains SAS/IML code to calculate the maximum leverage value inthe training set. The program reads in the test set, finds the leverage values for eachobservation and determines which, if any, observations are outside the training set space. Ifsome observations in the test set are outside the space, the program calculates theprediction error excluding these observations.

Program 3.7 Determine if the test set is in the training set space

proc reg data=soltsuse;model y=x1 x2 x3 x4 x5 x6 x7 x8 x9 x10 x11;output out=resds r=yresids;run;quit;

proc iml;use soltruse;read all var (’x1’:’x11’) into x;* Calculate the maximum leverage value of the training set;xtx=x‘*x;call eigen(evals,evecs,xtx);invxtx=evecs*diag(1/evals)*evecs‘;himax=max(vecdiag(x*invxtx*x‘));print himax[format=5.3];

Page 76: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 63

* Read in the test set and calculate the test set leverage values;* Determine which ones are in training set space;use soltsuse;read all var (’x1’:’x11’) into x;hi=vecdiag(x*invxtx*x‘);indi=choose(hi>himax,1,0);outindi=loc(indi);print outindi;use resds;read all var{yresids} into e;ein=e[loc(^indi),1];err=sqrt(sum(e##2)/(nrow(e)-ncol(x)));print err[format=5.3];quit;

Output from Program 3.7

Analysis of Variance

Sum of MeanSource DF Squares Square F Value Pr > F

Model 11 89.03923 8.09448 9.16 <.0001Error 37 32.69240 0.88358Corrected Total 48 121.73163

HIMAX

0.269

OUTINDI

6 40 41

ERR

0.928

The output analysis of variance table is contained in Output 3.7. The prediction error ofthe model, which is the square root of the Mean Square Error, calculated from the test setis

√0.8836 ≈ 0.94. The maximum leverage value in the training set is approximately 0.269,

so any observation in the test set whose leverage value is greater than this is not in thetraining set space. There were three observations in the test set that are outside thetraining set space (observations 6, 40 and 41). Excluding these observations results in aprediction error for the model of 0.928.

3.7 Using SAS Enterprise MinerAs an alternative to the procedures presented in the previous sections, model building canbe done using popular software, SAS Enterprise Miner. We will use SAS Enterprise Minerto build a model for the SOLUBILITY data set and make comparisons to the resultspresented in the previous sections.

To begin, we will use the basic SAS Enterprise Miner routines to input the solubilitydata using the drag and drop Input Data Source node. This particular node, the first onein Figure 3.4, allows the solubility data to be accessed by the software.

Page 77: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

64 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 3.4 SAS Enterprise Miner flow diagram

The second node in the Figure 3.4, the Data Partition node, provides an easy way topartition the data into training, test, and validation subsets. Table 3.1 below contains thesettings used in this example. We excluded a validation set so the results could becompared to the results obtained in Example 3.3.3.

Table 3.1 Settings Used in the Variable Selection Node

Setting value

Method Simple randomRandom seed 5164Training percentage 75%Validation percentage 0%Test percentage 25%

The output from running the Data Partition node is not included, but the procedureselected 128 observations for the training set and 42 observations for the test set.

The Variable Selection node, the third in Figure 3.4, was used to select descriptors thatare associated with the designated target variable, log-solubility. The rules used by thisnode are:

1. Compute the squared correlation for each descriptor with the target variable and thenassign the rejected role to those descriptors that have a value less than the squaredcorrelation criterion, 0.005.

2. Evaluate the remaining significant descriptors using a forward stepwise R2 regression.Descriptors that have a stepwise R2 improvement less than the threshold criterion,0.0005, are assigned the rejected role.

3. For binary descriptors, perform a logistic regression using the predicted values outputfrom the forward stepwise regression as the independent input.

The results from the variable selection process for the solubility data are shown below.It follows from the output that variables x5, x6, x7, x11, x15, and x18 were rejected becauseof a low R2 value.

Page 78: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 65

Output from the variable selection process

% # ofName Role Rejection Reason Missing Levels

X1 input 0 12X2 input 0 2X3 input 0 3X4 input 0 3X5 rejected Low R2 w/ target 0 11X6 rejected Low R2 w/ target 0 4X7 rejected Low R2 w/ target 0 5X8 input 0 122X9 input 0 127X10 input 0 116X11 rejected Low R2 w/ target 0 117X12 input 0 109X13 input 0 71X14 input 0 128X15 rejected Low R2 w/ target 0 8X16 input 0 49X17 input 0 125X18 rejected Low R2 w/ target 0 95X19 input 0 128X20 input 0 125X21 input 0 126

The fourth node in Figure 3.4 is the Data Set Attributes node. This node allows theuser to change certain features of the input data sets, which are the training and test setsafter variable selection. This node is needed to re-specify the target variable, or theresponse. The target variable was specified after running the Input Data Source node but isno longer specified after the Variable Selection node.

The next two nodes in Figure 3.4 are for constructing a model to predict solubility. Twomodels were fit to the training set, one using all the variables selected in the third nodeand the second using forward selection on the variables selected by the software.

The output below displays the output from fitting a linear model using all the variablesthe Variable Selection node did not reject. The output contains the analysis of variancetable, model fit statistics, and the parameter estimates along with the one-degree offreedom tests. The Root Mean Squared Error (RMSE) for this model is 0.6992. Thep-values from the one-degree of freedom tests indicate that some of the variables are notneeded in the model.

Output that shows solubility model performance

Analysis of Variance

Sum ofSource DF Squares Mean Square F Value Pr > F

Model 15 204.302215 13.620148 27.86 <.0001Error 112 54.750576 0.488844Corrected Total 127 259.052791

Page 79: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

66 Pharmaceutical Statistics Using SAS: A Practical Guide

Model Fit Statistics

R-Square 0.7887 Adj R-Sq 0.7603AIC -76.7030 BIC -70.1724SBC -31.0705 C(p) 16.0000

Analysis of Maximum Likelihood Estimates

StandardParameter DF Estimate Error t Value Pr > |t|

Intercept 1 4.0946 6.3716 0.64 0.5218x1 1 0.0973 0.0435 2.24 0.0273x16 1 0.1986 0.0631 3.15 0.0021x17 1 -18.6201 6.4189 -2.90 0.0045x19 1 -0.2266 5.7512 -0.04 0.9686x2 1 0.7320 0.2485 2.95 0.0039x20 1 -0.3103 0.1191 -2.60 0.0104x21 1 -0.2759 0.1118 -2.47 0.0151x4 1 0.6935 0.1936 3.58 0.0005x8 1 0.00276 0.00883 0.31 0.7554x9 1 0.3260 0.0984 3.31 0.0012x3 1 0.4099 0.2569 1.60 0.1135x12 1 -0.00290 0.00238 -1.22 0.2265x13 1 -0.00929 0.00322 -2.88 0.0047x14 1 -0.00695 0.00406 -1.71 0.0898x10 1 -0.00119 0.00229 -0.52 0.6043

The next output contains a subset of the output from the Regression node when forwardselection is used. The output displays a summary of forward selection, the analysis ofvariance table, model fit statistics, and parameter estimates along with the one-degree offreedom tests. The RMSE for this model is 0.7673 which is slightly higher than the modelusing all the variables. Therefore, the trade-off is complexity versus parsimony.

We decided to use the simpler model to obtain the model prediction error, which is theprediction error of the test data set. The Assessment node, the last node in Figure 3.4, isused to calculate the model prediction error. The RMSE for the test set is 1.0268.

Output that shows solubility model performance using forward selection

Summary of Forward Selection

Effect NumberStep Entered DF In F Value Pr > F

1 x8 1 1 89.36 <.00012 x16 1 2 94.15 <.00013 x3 1 3 13.48 0.00044 x2 1 4 4.28 0.0407

The selected model, based on the CHOOSE=AIC criterion, isthe model trained in Step 4. It consists of the following effects:

Intercept x16 x2 x8 x3

Page 80: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 3 Model Building Techniques in Drug Discovery 67

Analysis of Variance

Sum ofSource DF Squares Mean Square F Value Pr > F

Model 4 183.700468 45.925117 74.97 <.0001Error 123 75.352323 0.612621Corrected Total 127 259.052791

Model Fit Statistics

R-Square 0.7091 Adj R-Sq 0.6997AIC -57.8215 BIC -57.5483SBC -43.5614 C(p) 35.6797

Analysis of Maximum Likelihood Estimates

StandardParameter DF Estimate Error t Value Pr > |t|

Intercept 1 -0.0114 0.2261 -0.05 0.9599x16 1 0.3613 0.0362 9.97 <.0001x2 1 0.5384 0.2603 2.07 0.0407x8 1 -0.0187 0.00129 -14.48 <.0001x3 1 0.4819 0.1969 2.45 0.0158

3.8 SummaryThis chapter described statistical techniques used in drug discovery to facilitate modelbuilding. The techniques are useful for accelerating the pace of moving potential drugsthrough the development process. Procedures provided in this chapter deal withpartitioning relevant data into training and test sets, selecting potential variables to beused to construct the model, building the model, and making predictions.

The model building process described in the chapter is illustrated by using a real drugdiscovery data set to predict the solubility of various chemical compounds. The SAS code isprovided so that the interested reader can use these procedures on their own sets of data.

ReferencesBox, G.E.P., Hunter, J.S., Hunter, W.G. (1978). Statistics for Experimenters: Design, Innovation,

and Discovery. Second Edition. New York: Wiley.Breiman, L. (2001). “Statistical modeling: The two cultures.” Statistical Science. 16, 199–231.Efron, B., Tibshirani, R.J. (1998). An Introduction to the Bootstrap. Boca Raton, FL: CRC Press.Frank, I.E., Friedman, J.H. (1993). “A statistical view of some chemometrics regression tools.”

Technometrics. 35, 109–148.Furnival, G.M., Wilson, R.W. (1974). “Regression by leaps and bounds.” Technometrics. 16,

499–511.Golbraikh, A., Tropsha, A. (2002). “Predictive QSAR modeling based on diversity sampling of

experimental datasets for the training and test set selection.” Journal of Computer-AidedMolecular Design. 16, 357–369.

Golub, G.H., Van Loan, C.F. (1996). Matrix Computations. Third Edition. Baltimore, MD: TheJohn Hopkins Press.

Page 81: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

68 Pharmaceutical Statistics Using SAS: A Practical Guide

Hand, D. (1998). “Data mining: Statistics and more.” The American Statistician. 52, 112–118.Hastie, T., Tibshirani, R., Friedman, J. (2001). The Elements of Statistical Learning: Data Mining,

Inference and Prediction. New York: Springer-Verlag.Scott, D.W. (1979). “On optimal and data-based histograms.” Biometrika. 66, 3, 605–610.Two Crows Corporation (1999). Introduction to Data Mining and Knowledge Discovery. Third

Edition. Available at www.twocrows.com.

Page 82: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4

Statistical Considerations inAnalytical Method Validation

Bruno BoulangerViswanath DevanaryanWalthere DeweWendell Smith

4.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69

4.2 Validation Criteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73

4.3 Response Function or Calibration Curve. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .74

4.4 Linearity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83

4.5 Accuracy and Precision . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85

4.6 Decision Rule . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88

4.7 Limits of Quantification and Range of the Assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92

4.8 Limit of Detection. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .93

4.9 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93

4.10 Terminology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94

In the context of a pre-study validation of an analytical method, this chapter describessome advanced statistical methodologies available in the literature and provides SAS codeto calculate the validation criteria. Both linear and nonlinear methods are described withinthe calibration framework. In terms of validation criteria, i.e., when the calculatedconcentrations are available, criteria based on measurement error are utilized, and usefulgraphical representations are proposed.

4.1 IntroductionIn the pharmaceutical industry, analytical methods play a vital role in all experimentsperformed in the development of a drug product. If the quality of an analytical method isdoubtful, then the whole set of decisions based on those measures is questionable.

Bruno Boulanger is Manager, European Early Phase Statistics, Eli Lilly and Company, Belgium. Viswanath Devanaryan isAssociate Director, Biostatistics and Research Decision Sciences, Merck, USA. Walthere Dewe is Senior Research Scientist,Global Discovery and Development Statistics, Eli Lilly and Company, Belgium. Wendell Smith is Partner and SeniorStatistician, Bowsher Brunelle Smith (B2S Consulting), USA.

Page 83: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

70 Pharmaceutical Statistics Using SAS: A Practical Guide

Consequently, assessment of the quality of an analytical method is far more than astatistical challenge, it is a matter of good ethics and good business practices.

Many regulatory documents have been released in the pharmaceutical industry toaddress quality issues. These are primarily ICH and FDA documents. Those that arerelated to analytical and bioanalytical method validation (ICH, 1995, 1997; FDA, 2001)suggest that analytical methods must comply with specific acceptance criteria to berecognized as validated procedures. The primary aim of these documents is to requireevidence that the analytical methods are suitable for their intended use. Unfortunately,discrepancies exist among these documents with respect to the definition of acceptancecriteria, and limited guidance is provided for estimating the performance criteria.

In this chapter, background information will be provided on analytical methodvalidation concepts, and apparent inconsistencies will be addressed from a statisticalperspective. Statistical methods will be described for estimation of analytical performanceparameters, and decision criteria will be illustrated that are consistent with the concept ofa “good” analytical procedure. The impact of these methods on the design of experimentsneeded to obtain reliable estimates of the performance criteria will be considered. A majoremphasis of this chapter will be the use of SAS programs to illustrate the computation ofassay performance parameters, with limited discussion about the philosophy of the assayvalidation purpose or practices.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

4.1.1 Method Classification Based on Data TypesThe ultimate goal of an analytical method or procedure is to measure accurately aquantity, such as the concentration of an analyte, or to measure a specific activity, as forexample for a biomarker. However, many assays such as cell-based and enzyme activitybiomarker assays may not be very sensitive, may lack precision, and/or may not offerdefinitive reference standards. Assays based on physicochemical (such as chromatographicmethods) or biochemical (such as ligand binding assays) properties of an analyte assumethat these quantifiable characteristics are reflective of the quantity, concentration, orbiological activity of the analyte. For the purpose of analytical validation, we will followthe recently proposed classifications for assay data by Lee et al. (2003). Theseclassifications, summarized below, provide a clear distinction with respect to the analyticalvalidation practice and requirements.

Qualitative methods generate data which do not have a continuous proportionalityrelationship with the amount of analyte in a sample; the data are categorical in nature.Data may be nominal such as a present/absent call for a gene or gene product.Alternatively, data might be ordinal in nature, with discrete scoring scales (e.g., 1 to 5 or−, +, + + +) such as for immuno-histochemistry assays or Fluorescence In SituHybridization (FISH).

Quantitative methods are assays where the response signal has a continuous relationshipwith the quantity or activity of the analyte. These responses can therefore be described bya mathematical function. Inclusion of reference standards at discrete concentrations allowsthe quantification of sample responses by interpolation. The availability of a well-definedreference standard may be limited, or may not be representative of the in vivopresentation, so quantification may not be absolute. To that end, three types ofquantitative methods have been defined:

• A definitive quantitative assay uses calibrators fit to a known model to provide absolutequantitative values for unknown samples. Typically, such assays are possible only wherethe analyte is not endogenous. An example of this is a small molecule drug.

Page 84: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 71

• A relative quantitative assay is similar in approach, but generally involves themeasurement of endogenously occurring analytes. In this case, even a “zero” or blankcalibrator may contain some amount of analyte, and quantification can only be donerelative to this “zero” level. Examples of this include immunoassays for cytokines suchas sTNFRII, or gene expression assays, e.g., Reverse Transcriptase-Polymerase ChainReaction (RT-PCR).

• A quasi-quantitative assay does not involve the use of calibrators, mostly due to the lackof suitable reference material, so the analytical result for a test sample is reported onlyin terms of the assay signal (e.g., optical density in ELISA).

This chapter deals with the assessment of definite and relative quantitative assays. Afull discussion of quasi-quantitative and qualitative assays and statistical considerationsthereof is beyond the scope of this chapter. A good reference on the analytical validation ofa typical quasi-quantitative assay is a white paper on immunogenicity by Mire-Sluis et al.(2004).

4.1.2 Objective of an Analytical MethodThe objective of a definite and relative quantitative analytical method is to be able toquantify as accurately as possible each of the unknown quantities that the laboratory willhave to determine. In other words, what all analysts expect from an analytical procedure isthat the difference between the measurement or observation (X) and the unknown “truevalue” μT of the test sample be small or inferior to an acceptance limit λ:

−λ < X − μT < λ ⇐⇒ |X − μT | < λ. (4.1)

The acceptance limit λ can be different depending on the requirements of the analystand the objective of the analytical procedure. The objective is linked to the requirementsusually admitted by the practice (e.g., 1% or 2% on bulk, 5% on pharmaceuticalspecialties, 15% for biological samples. Acceptance limits vary in clinical applicationsdepending on factors such as the physiological variability and the intent of use).

4.1.3 Objective of the Pre-Study Validation PhaseThe aim of the pre-study validation phase is to generate information to guarantee that theanalytical method will provide, in routine use, measurements close to the true value(DeSilva et al., 2003; Findlay, 2001; Hubert et al., 2004; Smith and Sittampalam, 1998;Finney, 1978) without being affected by other elements present in the sample. In otherwords, the validation phase should demonstrate that the inequality described inEquation (4.1) holds for a certain proportion of the sample population.

The difference between the measurement X and its true value is a sum of a systematicerror (bias or trueness) and a random error (variance or precision). The true values of theseparameters are unknown but they can be estimated based on the validation experiments.The reliability of these estimates depends on the adequacy of these experiments (design,size).

Consequently, the objective of the validation phase is to evaluate whether, given theestimates of bias and variance, the expected proportion of measures that will fall withinthe acceptance limits is greater than a predefined level, say, β:

Eμ,σ (P [|X − μT | < λ|μM , σM ]) ≥ β. (4.2)

Although Equation (4.2) cannot be solved exactly within a frequentist framework,Section 4.6 will discuss approximate solutions that can be used in practice.

Page 85: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

72 Pharmaceutical Statistics Using SAS: A Practical Guide

4.1.4 Classical Design in Pre-Study ValidationExperiments performed during pre-study validation are designed to mimic the processesand practices to be followed during routine application of a method. All aspects of theanalytical method should be taken into account, such as the lot of a solvent, operator,preparation of samples, etc. If measurements generated under these “simulated” conditionsare acceptable (see Section 4.6) then the method will be declared valid for routine use.Usually, two sets of samples will be prepared for simulating the real process: calibrationand validation samples.

• Calibration samples (CS) must be prepared according to the protocol that will befollowed during routine use, i.e., the same operational mode, the same number ofconcentration levels for the standard curve, and the same number of repetitions at eachlevel.

• Validation samples (VS) must be prepared in the sample matrix when applicable. In thevalidation phase, they mimic the unknown samples that the analytical procedure willhave to quantify in routine use. Each validation standard should be preparedindependently, in order to have realistic estimates of the variance components.

The minimum design of a pre-study validation phase is at least two replicates per run orseries in a minimum of three runs. However, it is highly recommended to consider at leastsix runs in order to have a good estimate of the between-run variance. The number of runsand replicates to perform at each concentration level to demonstrate that an analyticalprocedure is valid could be estimated (by simulations) and depends, of course, on theinherent but unknown properties of the analytical procedure itself. The more variable themethod, the more experiments are necessary.

Table 4.1 displays the minimal sample size for r runs and s replicates per run for 10%acceptance limits (the table was computed via simulations, assuming a potential small biasof 2%). It is clear that the number of runs increases with increasing between-run variance.The higher number of runs can be compensated for by more replicates per runs, but thisleads to a larger total number of experiments (rs). Also, as expected when the sum of bias(2%) and the within-run and between-run variances become greater than 10%, it becomesunlikely that the method will ever be validated for such acceptance limits. Moredevelopment in the laboratory is required to achieve this objective. The reproducibilitythat requires between-laboratory experiments will not be discussed in this chapter.

Table 4.1 Minimal Sample Size for r Runs and s Replicates per Run for 10% Acceptance Limits

Within-run variance1% 2% 3% 4% 5%

Between-run

variance r s r s r s r s r s

1% 4 3 4 3 4 3 4 4 5 95 3 5 3 5 3 5 4 6 7

2% 4 3 4 3 4 3 4 6 8 95 3 5 3 5 3 5 6 9 7

3% 4 4 4 6 5 6 7 105 3 5 3 6 5 8 9

4% 7 10 9 88 7 10 6

Page 86: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 73

4.1.5 Example: A Sandwich ELISA AssayA sandwich ELISA assay, optimized by statistical design of experiments and validated atLilly Research Laboratories, will be used throughout this chapter for illustration purposes.This data set is available on the book’s companion Web site.

The objective of this assay was to quantify a protein used as a biomarker in neurologicaldisease therapeutic research. The ELISA consisted of incubation of samples on platespre-coated with a capture antibody specific to the protein of interest followed byimmunological detection of the specific bound protein by an enzyme conjugate andmeasurement (optical density) of the colored product. In order to validate this assay,calibration standards and validation samples were prepared in appropriate matrices fromstock protein solution via serial dilution and then tested in triplicate. The procedure wasfollowed for four independent runs with two plates per run over four days.

4.2 Validation CriteriaThe main validation criteria widely recommended by various regulatory documents (ICH,FDA, European Union) and commonly used in analytical laboratories are:

• Specificity-selectivity.• Response function (calibration curve).• Linearity.• Precision (repeatability and intermediate precision).• Accuracy (trueness).• Measurement error (total error).• Limit of detection (LOD).• Limit of quantification (LOQ).• Assay range.• Sensitivity.

In addition, according to the domains concerned, other specific criteria can be required:

• Analyte stability.• Recovery.• Effect of the dilution.

A full validation is necessary for an analytical procedure to pass from the developmentphase to the phase of routine analysis. The validation step is not only necessary but alsorequired at the time specifications (tests and acceptance limits) are set up for an activeingredient or a finished product.

The validation criteria mentioned above must be established, insofar as possible, in thesame matrix as that of samples to be analyzed. Every new analytical procedure will have tobe validated for each type of matrix (e.g., for each type of biological fluid and for eachanimal species). Nevertheless, the definition of a matrix depends on analyst responsibility.Some matrix regrouping, generally admitted by the profession for an application domaingiven, can be performed.

This section will focus only on the main criteria that apply to most, if not all, analyticalmethods and that must be adequately estimated and documented for ensuring complianceto regulations. The criteria that are very analytical, but that have no particular statisticalcontent, will be defined here, but will not be discussed.

Page 87: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

74 Pharmaceutical Statistics Using SAS: A Practical Guide

4.2.1 Specificity-SelectivityThe specificity of an analytical procedure is the ability to unequivocally assess the analytein the presence of components that may be expected to be present. Usually, the analystmust demonstrate that the measured result is directly related to the analyte or product ofinterest and that other aspects in the sample, such as the matrix, do not interfere with thesignal or measurement. For example, selectivity of a chromatographic method is verifiedtypically by showing that the product of interest is clearly separated from all otherproducts.

4.3 Response Function or Calibration CurveThe response function for an analytical procedure is the existing relationship, within aspecified range, between the response (signal, e.g., area under the curve, peak height,absorption) and the concentration (quantity) of the analyte in the sample. The calibrationcurve should be described preferably by a simple monotonic response function that givesaccurate measurements. Note that the response function is frequently confused with thelinearity criteria. However, the latter criterion refers to the relationship between thequantity introduced and the quantity back-calculated from the calibration curve (seeSection 4.4). Because of the confusion, it is common to see laboratory analysts try veryhard to ensure that the response function is linear in the classical sense, i.e., a straight line.Not only is this not required, but it is often irrelevant and can lead to large errors inmeasured results (e.g., for ligand binding assays). A significant source of bias andimprecision in analytical measurements can be the choice of the statistical model for thecalibration curve.

4.3.1 Computational AspectsStatistical models for calibration curves can be either linear or nonlinear in theirparameter(s). The choice between these two families of models will depend on the type ofmethod and/or the range of concentrations of interest. If the range is very narrow, anunweighted linear model may suffice, while a larger range may require a more advancedand weighted model. High Performance Liquid Chromatography (HPLC) methods areusually linear while immunoassays are typically nonlinear. Weighting may be important forboth methods because a common feature for many analytical methods is that the varianceof the signal is a function of the level or quantity to be measured.

Methodologies for fitting linear and nonlinear models generally require different SASprocedures. For both model types, curves are fit by finding values for the model parametersthat minimize the sum of squares of the distances between observations and the fittedcurve. For linear models, parameter estimates can be derived analytically while this is notthe case for many nonlinear models. Consequently, iterative procedures are often requiredto estimate the parameters of a nonlinear model. In this section, both linear and nonlinearmodels will be considered.

In case of heterogeneous variances of the signal across the concentration range, it isrecommended that observations be weighted when fitting a curve. If observations are notweighted, an observation more distant to the curve than others has more influence on thecurve fit. As a consequence, the curve fit may not be good where the variances are smaller.Weighting each term of the sum of squares is frequently used to solve this problem, wherethis can be viewed as minimizing the relative distances instead of minimizing the actualdistances. When replicates are present at each concentration level, it is often better to fitthe model to their average/median response values. Regardless of model type, it is assumedthat all observations fit to a model are completely independent. In reality, replicates areoften not independent for many analytical procedures because of the steps followed inpreparation and analysis of samples. In such cases, replicates should not be used separately.

Page 88: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 75

Models are typically applied on either a linear scale or log scale of the assay signaland/or the calibrator concentrations. The linear scale is used in case of homogeneousvariance across the concentration range, and the log scale is often more appropriate whenvariance increases with increasing response.

4.3.2 Linear and Polynomial ModelsMost commonly used types of polynomial models include simple linear regression (with orwithout an intercept) and quadratic regression models.

As an illustration, consider a subset of the ELISA CS data set (Plate A, Series 1) thatincludes three replicate measurements at each of eight concentration levels (the ELISA CSdata set is provided on the book’s web site). The selected measurements are included in theCALIB data set shown below in Program 4.1. The CONCENTRATION variable is theconcentration value and the rep1, rep2, and rep3 variables represent the three replicates.Program 4.1 uses the MIXED procedure to fit a linear model to the data collected in thestudy. The model parameters are estimated using the restricted maximum likelihoodmethod, which is equivalent to the ordinary least square method when the data arenormally distributed. The SOLUTION option requests parameter estimates which are thensaved to a SAS data set specified in the ODS statement. The OUTPREDM option is usedto save the predicted signal values from the fitted linear model to another SAS data set.The WEIGHT statement is used to assign weights to the individual measurements (the Wvariable represents the weight). For example, the weights can be defined as the inverse ofthe signal level or the inverse of the squared signal. In this example, the weights are definedusing the following formula: w = 1/s1.3, where s is the signal level. The choice of theexponent (e.g., 1.3) depends on the relationship between the signal’s variability and itsaverage level.

Program 4.1 Fitting a simple linear regression model using PROC MIXED

data calib;set elisa_cs;if plate=’A’ and series=1;array y(3) rep1 rep2 rep3;do j=1 to 3;

signal=y(j);w=1/signal**1.3;

output;end;

proc mixed data=calib method=reml;model signal=concentration/solution outpredm=predict;weight w;ods output solutionf=parameter_estimates;

proc sort data=predict;by concentration;

axis1 minor=none label=(angle=90 ’Signal’) order=(0 to 4 by 1);axis2 minor=none label=(’Concentration’) logbase=10 logstyle=expand;symbol1 value=none i=join color=black line=1 width=3;symbol2 value=dot i=none color=black height=5;proc gplot data=predict;

plot (pred signal)*concentration/overlay frame haxis=axis2 vaxis=axis1;run;quit;

Page 89: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

76 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 4.1

Solution for Fixed Effects

StandardEffect Estimate Error DF t Value Pr > |t|

Intercept 0.3684 0.02850 22 12.93 <.0001concentration 0.005802 0.000339 22 17.12 <.0001

Output 4.1 lists the parameter estimates generated by PROC MIXED and associatedp-values. Figure 4.1 displays the fitted regression line. It is obvious that the linear modelprovides a poor fit to the data.

Figure 4.1 Fit of the linear model

Sig

nal

0

1

2

3

4

Concentration

1 10 100 1000

While Program 4.1 focuses on a simple linear regression model with an intercept, onecan consider a linear regression model without an intercept, which is requested using thefollowing MODEL statement:

model signal=concentration/solution noint;

or a quadratic regression model:

model signal=concentration concentration*concentration/solution;

In addition, the BY statement can be included in PROC MIXED to fit a calibrationcurve within each run.

4.3.3 Nonlinear Models (PROC NLIN)As briefly mentioned in the introduction of this section, to fit a nonlinear model, one needsto rely on iterative methods. These methods begin with an initial set of parameter valuesfor the model of interest and update the parameter values at each step in order to improvethe fit. The iterative process is stopped when the fit can no longer be improved.

Nonlinear models frequently used in curve calibration include the 4-parameter logisticregression, 5-parameter logistic regression and power model. Consider, for example, the4-parameter logistic regression model:

y = f(x) = β1 +β2 − β1

1 + (x/β3)β4,

Page 90: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 77

where β1, β2, β3, and β4 are the top asymptote, bottom asymptote, and concentrationcorresponding to half distance between β1 and β2, and the slope, respectively.

Both the NLIN and NLMIXED procedures can be used to fit such models. Except forthe fact that they both optimize a function of interest, they do not work in the samemanner. PROC NLIN fits nonlinear models by minimizing the error sum of squares and itcan handle only models with fixed effects. PROC NLMIXED enables us to fit models withfixed and random effects by maximizing an approximation to the likelihood functionintegrated over the random effects. In this context, PROC NLMIXED is used only inmodels with fixed effect, and thus the problem of integration is avoided.

Program 4.2 relies on PROC NLIN to model the relationship between concentration andsignal levels in the subset of the ELISA CS data set. PROC NLIN supports severaliterative methods for minimizing the error sum of squares (they can be specified using theMETHOD statement). The more robust methods are GAUSS, NEWTON, andMARQUARDT. The default method is GAUSS, but the most commonly used isMARQUARDT.

Program 4.2 analyzes the CALIB data set created in Program 4.1. The initial values ofthe four parameters are specified in the PARAMETERS statement and the WEIGHTstatement is used to weight the observations when fitting the model.

Program 4.2 Fitting a 4-parameter logistic regression model using PROC NLIN

proc nlin data=calib method=marquardt outest=parameter_estimates;parameters top=3 bottom=0.2 c50=250 slope=1;model signal=top+(bottom-top)/(1+(concentration/c50)**slope);_weight_=w;output out=fitted_values predicted=pred;

proc sort data=fitted_values;by concentration;

axis1 minor=none label=(angle=90 ’Signal’) order=(0 to 4 by 1);axis2 minor=none label=(’Concentration’) logbase=10 logstyle=expand;symbol1 value=none i=join color=black line=1 width=3;symbol2 value=dot i=none color=black height=5;proc gplot data=fitted_values;

plot (pred signal)*concentration/overlay frame haxis=axis2 vaxis=axis1;run;quit;

Output from Program 4.2

Sum of Mean ApproxSource DF Squares Square F Value Pr > F

Model 3 9.5904 3.1968 6181.88 <.0001Error 20 0.0103 0.000517Corrected Total 23 9.6008

Approx Approximate 95% ConfidenceParameter Estimate Std Error Limits

top 3.6611 0.1186 3.4138 3.9085bottom 0.3198 0.00681 0.3056 0.3340c50 219.9 13.9168 190.9 249.0slope 1.2611 0.0391 1.1795 1.3427

Page 91: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

78 Pharmaceutical Statistics Using SAS: A Practical Guide

Output 4.2 shows that the overall F value is very large while the standard errors of thefour parameter estimates are small. This suggests that the model’s fit was excellent (seealso Figure 4.2). It is important to note that one sometimes encounters convergenceproblems and it is helpful to examine the iteration steps included in the output. If theiterative algorithm does not converge, it is prudent to explore different sets of initial valuesthat can be obtained by a visual inspection of the raw data.

Figure 4.2 Fit of the 4-parameter logistic regression model

Sig

nal

0

1

2

3

4

Concentration

1 10 100 1000

4.3.4 Nonlinear Models (PROC NLMIXED)PROC NLMIXED supports a large number of iterative methods for fitting nonlinearmodels. Unfortunately, there is no general rule for choosing the most appropriate method.The choice is problem-dependent and, most of the time, one needs to select the iterativemethod by trial and error (see PROC NLMIXED documentation for generalrecommendations).

Note that the METHOD option in PROC NLMIXED does not specify the optimizationmethod as in PROC NLIN, but rather the method for approximating the integral of thelikelihood function over the random effects. The TECHNIQUE option is used in PROCNLMIXED to select the optimization method. Program 4.3 fits a 4-parameter logisticregression model to the concentration/signal data from the CALIB data set created inProgram 4.1. The program uses the Newton-Raphson method with ridging as anoptimization method (NRRIDG) in PROC NLMIXED. Other TECHNIQUE optionsinclude NEWRAP (Newton-Raphson optimization combining a line-search algorithm withridging) or QUANEW (quasi-Newton). Since no random effects are included in this model,the METHOD option is not used in this example.

PROC NLMIXED does not allow the fitting of weighted regression models; however, itallows us to specify a variance function. The most popular variance function in nonlinearcalibration is the power of the mean (O’Connell, Belanger, and Haaland, 1993). To specifythis function in Program 4.3, we introduce the VAR and THETA parameters:

model signal~normal(expect,(expect**theta)*var);

Here VAR is the residual variance at baseline, and THETA defines the rate at whichthis variance changes with the predicted concentration (EXPECT variable). The

Page 92: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 79

FITTED VALUES data set contains the fitted value for each concentration value as well asthe corresponding residual.

Program 4.3 Fitting a 4-parameter logistic regression model using PROC NLMIXED

proc nlmixed data=calib technique=nrridg;parms top=3 bottom=0.2 c50=250 slope=1 theta=1 var=0.0001;expect=top+(bottom-top)/(1+(concentration/c50)**slope);model signal~normal(expect,(expect**theta)*var);predict top+(bottom-top)/(1+(concentration/c50)**slope) out=fitted_values;run;

Output from Program 4.3

Parameter Estimates

StandardParameter Estimate Error DF t Value Pr > |t| Alpha Lower Upper

top 3.6584 0.1213 24 30.16 <.0001 0.05 3.4081 3.9087bottom 0.3205 0.006942 24 46.18 <.0001 0.05 0.3062 0.3349c50 219.50 14.2383 24 15.42 <.0001 0.05 190.12 248.89

slope 1.2628 0.04151 24 30.42 <.0001 0.05 1.1771 1.3485theta 1.2879 0.4150 24 3.10 0.0049 0.05 0.4313 2.1445var 0.000434 0.000130 24 3.35 0.0027 0.05 0.000167 0.000701

Output 4.3 shows that the parameter estimates and their approximate standard errorsproduced by PROC NLMIXED are very close to those displayed in the PROC NLINoutput (Output 4.2). This is partly due to the fact that the estimated THETA parameter(1.2879) is very close to the exponent used in the weighting scheme in PROC NLIN. Thefitted calibration curve is very similar to the calibration curve displayed in Figure 4.2.

4.3.5 Precision Profile for Immuno-AssaysAfter a calibration curve and weighting model have been chosen, a precision profile may beemployed to characterize the precision of the back-calculated concentrations for unknowntest samples using this calibration curve. The precision profile is a plot of thecoefficient-of-variation (CV) of the calibrated concentration versus the true concentrationon a log scale. Ideally, the calculated standard error of the calibrated concentration musttake into account both the variability in the calibration curve and variability in the assayresponse. Wald’s method is generally recommended for computing these standard errors(Belanger, Davidian and Giltinan, 1996) and the resulting coefficient-of-variation is givenby:

CV(x0) =100x0

[(∂f−1(y0, β)

∂y

)σ2y2θ

0

m+

(∂f−1(y0, β)

∂y

)′

Σ(β)

(∂f−1(y0, β)

∂y

)]1/2

,

where m is the number of replicates and Σ(β) is the covariance matrix of the parameterestimates β.

As an illustration, Program 4.4 computes a precision profile for a 5-parameter logisticmodel:

y = f(x) = β1 +β2 − β1

[1 + (x/β3)β4 ]γ.

Page 93: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

80 Pharmaceutical Statistics Using SAS: A Practical Guide

The γ parameter is known as the asymmetry factor and, when it is set to 1, this model isequivalent to a 4-parameter logistic model fitted in Program 4.3. In fact, Program 4.4 fitsthis special case of the 5-parameter logistic model by forcing the γ parameter to be 1 (notethe restrictions on this parameter in the BOUNDS statement). A general 5-parameterlogistic model can be obtained by removing these constraints. Further, Program 4.4 usesthe IML procedure to calculate the precision profile (in general, it is easier to use thematrix language instead of DATA steps to calculate the two terms under the square root).The covariance matrix is extracted from PROC NLMIXED and is imported into PROCIML. The computed response profile is displayed in Figure 4.3.

Program 4.4 Computation of the precision profile for a 4-parameter logistic regression model using PROCNLMIXED

proc nlmixed data=calib technique=nrridg;parms top=3 bottom=0.2 c50=250 slope=1 theta=1 var=0.0001 g=1;bounds g>=1, g<=1, theta>=0, var>0; * Constraints on model parameters;

expect=top+(bottom-top)/((1+(concentration/c50)**slope)**g);model signal~normal(expect,(expect**theta)*var);predict top+(bottom-top)/((1+(concentration/c50)**slope)**g) out=fitted_values;ods output ParameterEstimates=parm_est_repl;

ods output CovMatParmEst=cov_parm;* Data set containing parameter estimates;data b;

set parm_est_repl;where parameter in (’top’,’bottom’,’c50’,’slope’);keep estimate;

* Data set containing the covariance matrix of the parameter estimates;data covb;

set cov_parm;where parameter in (’top’,’bottom’,’c50’,’slope’);

keep top bottom c50 slope;proc sql noprint;

select distinct estimate into: sigma_sq from parm_est_repl where parameter=’var’;

select distinct estimate into: theta from parm_est_repl where parameter=’theta’;select min(concentration) into: minconc from calib where concentration>0;select max(concentration) into: maxconc from calib;

run;quit;

%let m=3; * Number of replicates at each concentration level;proc iml;

* Import the data sets;use b; read all into b;use covb; read all into covb;

* Initialize matrices;y=j(101,1,0);h=j(101,1,0);

hy=j(101,1,0);hb=j(101,4,0);varx0=j(101,1,0);

pp=j(101,1,0);top=b[1];bottom=b[2];c50=b[3];

slope=b[4];

Page 94: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 81

* Calculate the precision profile;do i=1 to 101;

h[i,1]=10**(log10(&minconc)+(i-1)*(log10(&maxconc)-log10(&minconc))/100);

y[i,1]=top+(bottom-top)/(1+(h[i,1]/c50)**slope);hy[i,1]=h[i,1]*(top-bottom)/(slope*(bottom-y[i,1])*(y[i,1]-top));hb[i,1]=h[i,1]/(slope*(y[i,1]-top));

hb[i,2]=h[i,1]/(slope*(bottom-y[i,1]));hb[i,3]=h[i,1]/c50;hb[i,4]=-h[i,1]*log((bottom-y[i,1])/(y[i,1]-top))/(slope**2);

varx0[i,1]=((hy[i,1]**2)*&sigma_sq*(y[i,1]**(2*&theta))/&m)+hb[i,]*covb*hb[i,]‘;pp[i,1]=100*sqrt(varx0[i,1])/h[i,1];

end;create plot var{h hy y pp};

append;quit;

axis1 minor=none label=(angle=90 ’CV (%)’) order=(0 to 30 by 10);

axis2 minor=none label=(’Concentration’) logbase=10 logstyle=expand;symbol1 value=none i=join color=black line=1 width=3;proc gplot data=plot;

plot pp*h/frame haxis=axis2 vaxis=axis1 vref=20 lvref=34 href=4.2 lhref=34;run;quit;

Figure 4.3 Precision profile based on a 4-parameter logistic regression model

CV

(%

)

0

10

20

30

Concentration

1 10 100 1000

It can be seen in Figure 4.3 that, for concentration levels above 4 μM, the fitted modelis a priori able to quantify with a precision better than 20% on a CV scale. Below thisthreshold, the variability of the back-calculated measurements explodes as expectedbecause of the (low) asymptote effect. The precision achieves its maximum (approximately2-3% on a CV scale) around the C50 value (220 μM).

The estimates of quantification limits from precision profiles are “optimistic” becausethey are based on only the calibration curve data themselves. These limits do not take intoaccount matrix interference, cross-reactivity, operational factors, etc. However, these limitsserve as a useful screening tool before beginning the pre-study validation exercise. Since thepre-study validation package encompasses several other sources of variability as well, if thequantification limits from a precision profile are not satisfactory, then almost definitely, thequantification limits derived from a rigorous pre-study validation package will not besatisfactory. In this case, it will be worth going back to the drawing board and furtheroptimizing the assay protocol before proceeding to the pre-study validation phase.

Page 95: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

82 Pharmaceutical Statistics Using SAS: A Practical Guide

4.3.6 Back-Calculated Quantities or Inverse PredictionsOnce a calibration curve is fitted, concentrations of the samples of interest are calculatedby inverting the estimated calibration function. In a pre-study validation, the calibrationcurves are fitted separately for each run and the validation samples are calculated using thecalibration curve for the same run. The resulting data set consists of different concentrationlevels and, at each level, there are multiple runs and replicates within each run. Most of thetime, the number of runs and the number of replicates are the same for all concentrationlevels. Inverse functions for widely used response functions are shown in Table 4.2.

For example, Program 4.5 calculates the concentrations of the validation samples(ELISA VS data set available on the book’s companion Web site) from the 4-parameterlogistic regression model fitted by series and by plate as described in Program 4.3.

Program 4.5 Concentration calculation based on a 4-parameter logistic regression model

data calib;set elisa_cs;array y(3) rep1 rep2 rep3;do j=1 to 3;

signal=y(j);output;end;

proc datasets nolist;delete parm_est;

%macro calib(series, plate);proc nlmixed data=calib technique=nrridg;

parms top=3 bottom=0.2 c50=250 slope=1 theta=1 var=0.0001;expect=top+(bottom-top)/(1+(concentration/c50)**slope);model signal~normal(expect,(expect**(theta*2))*var);where series=&series and plate=&plate;ods output ParameterEstimates=parm_est_tmp;

data parm_est_tmp;set parm_est_tmp;series=&series;plate=&plate;

proc append data=parm_est_tmp base=parm_est;run;

%mend;%calib(series=1, plate=’A’);%calib(series=1, plate=’B’);%calib(series=2, plate=’A’);%calib(series=2, plate=’B’);%calib(series=3, plate=’A’);%calib(series=3, plate=’B’);%calib(series=4, plate=’A’);%calib(series=4, plate=’B’);data valid;

set elisa_vs;array y(3) rep1 rep2 rep3;do j=1 to 3;

signal=y(j);output;

end;proc sort data=valid;

by series plate;

Page 96: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 83

proc transpose data=parm_est out=parm_est_t;var estimate;id parameter;idlabel parameter;by series plate;

data calc_conc;merge valid parm_est_t;by series plate;drop _name_ theta var;

data calc_conc;set calc_conc;if plate=’A’ then run=series;else run=series+4;calc_conc=c50*((((bottom-top)/(signal-top))-1)**(1/slope));run;

Table 4.2 Inverse Functions for Widely Used Response Functions

Back-calculated value x∗

Response function using the inverse function

Y = βX x∗ = y/β

Y = β0 + β1X x∗ = (y − β0)/β1

Y = β0 + β1X + β2X2 x∗ =

(−β1 +

√β2

1 − 4β2(β0 − y))

/2β2

Y = β1 + β2 − β11 + (X/β3)β4

x∗ = β3

(β2 − β1

y − β1− 1

)−1/β4

4.4 LinearityThe linearity of an analytical procedure is defined in terms of its ability to obtain resultsdirectly proportional to the concentrations (quantities) of the analyte in the sample withina defined range (ICH, 1995). It is important to note that the linearity criterion is appliedto the results, i.e., back-calculated quantities or concentrations, rather than the responsesignals or instrument response as a function of the dose or quantities.

To illustrate this concept, Program 4.6 performs a linearity assessment by comparingthe original concentrations from the ELISA VS data set to the correspondingback-calculated concentrations obtained in Program 4.5. This program uses theCALC CONC data set created in Program 4.5. The results are displayed in Figure 4.4.

Program 4.6 Linearity assessment based on a 4-parameter logistic regression model

proc sql;create table linprof asselect concentration, calc_conc, ’t1’ as type

from calc_conc unionselect distinct concentration, concentration*0.7 as calc_conc, ’t2’ as type

from calc_conc unionselect distinct concentration, concentration*1.3 as calc_conc, ’t3’ as type

from calc_conc union

Page 97: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

84 Pharmaceutical Statistics Using SAS: A Practical Guide

select distinct concentration, concentration as calc_conc, ’t4’ as typefrom calc_conc;

run;quit;

proc sort data=linprof;by type concentration;

axis1 label=("Nominal concentration") order=(0 to 600 by 200);axis2 label=(angle=90 "Calculated concentration") order=(0 to 600 by 200);symbol1 value=circle i=none color=black height=5;symbol2 value=none i=join color=black line=34 width=3;symbol3 value=none i=join color=black line=34 width=3;symbol4 value=none i=join color=black line=1 width=3;proc gplot data=linprof;

plot calc_conc*concentration=type/haxis=axis1 vaxis=axis2 nolegend;run;quit;

Figure 4.4 Linearity assessment based on a 4-parameter logistic regression model

Cal

cula

ted

conc

entr

atio

n

0

200

400

600

Nominal concentration

0 200 400 600

Figure 4.4 plots the raw concentrations versus the corresponding back-calculatedconcentrations. The solid line is the identity line and, if the method is reasonably unbiased,all data points are expected to cluster around this line. The two dotted lines represent the[−30%, 30%] acceptance limits drawn on an absolute scale. If the precision of a method issatisfactory, most data points should lie between the two lines.

Figure 4.4 displays the linearity of the results for the data obtained using the calibrationcurve of Figure 4.2. The contrast between those two figures highlights clearly the contrastbetween the concepts of linearity of the results and response function of the signals. Whenchromatographic methods with narrow ranges are envisaged, both the response functionand linearity will look linear but, except for this specific case, the two graphs will lookdifferent.

Page 98: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 85

4.5 Accuracy and Precision

4.5.1 Accuracy (Trueness)The accuracy (the preferred term is trueness) of an analytical procedure, according to ICHand related documents, expresses the closeness of agreement between the mean valueobtained from a series of measurements and the value which is accepted either as aconventional true value or an accepted reference value (e.g., international standard,standard from a pharmacopoeia). It is a measure of the systematic error of test resultsobtained by the analytical method from its theoretical true/reference value. The measureof trueness is generally expressed in terms of recovery and absolute/relative bias.

Note that “accuracy” is synonymous with bias or trueness only within thepharmaceutical set of regulations covered by ICH (and related national documentsimplementing ICH Q2A and Q2B). Outside the pharmaceutical industry, e.g., in industriescovered by the International Organization for Standardization or National Committee forClinical Laboratory Standards guidelines (food, chemistry, clinical biology, and otherindustries), “accuracy” refers to the total error, i.e., the sum of trueness and precision.

4.5.2 PrecisionThe precision of an analytical procedure is defined by the closeness of agreement (usuallyexpressed as standard deviation or coefficient of variation) between a series ofmeasurements obtained from multiple sampling of the same homogeneous sample(independent assays) under the prescribed conditions. The term “independent results”means that the results are obtained and prepared the same way that unknown samples willbe quantified and prepared.

Precision provides information on random errors and can be evaluated at three levels:repeatability, intermediate precision (within laboratory), and reproducibility (betweenlaboratories). The precision represents the distribution of the random errors only and is notrelated to the true or specified value. A measure of precision is calculated from thestandard deviation of the results.

Quantitative measures of precision depend in a critical manner on stipulated conditions.One can distinguish among the following three conditions:

• Repeatability. Repeatability expresses the precision under conditions where the resultsof independent assays are obtained by the same analytical procedure on identicalsamples in the same laboratory, with the same operator, using the same equipmentduring a short interval of time. It is estimated by the within-series variance component.

• Intermediate precision. Intermediate precision expresses the precision underconditions where the results of independent assays are obtained by the same analyticalprocedure on identical samples in the same laboratory, with different operators, usingdifferent equipment during a given time interval. It is estimated by the sum ofwithin-series and between-series variance components. Intermediate precision isrepresentative of the total random error for a single measurement within a laboratory,whatever the day or series.

• Reproducibility. Reproducibility expresses the precision under conditions where theresults are obtained by the same analytical procedure on identical samples in differentlaboratories, with different operators, using different equipment. It is estimated by thesum of within-series, between-series and between-laboratories variance components.

4.5.3 Total Error or Measurement ErrorThe measurement error of an analytical procedure is related to the closeness of agreementbetween the value found and the value that is accepted either as a conventional true value

Page 99: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

86 Pharmaceutical Statistics Using SAS: A Practical Guide

or an accepted reference value. The closeness of agreement observed is based on the sum ofthe systematic and random errors; in other words, the total error linked to the result.Consequently, the measurement error is the expression of the sum of the trueness andprecision, i.e., the total error.

As shown below, the observation X is a result of the true sample value μT , the method’sBias (estimated by the mean of many results) and Precision (estimated by the standarddeviation or, in most cases, the Intermediate Precision). Equivalently, the differencebetween an observation X and the true value is the sum of the systematic and randomerrors, i.e., Total Error or Measurement Error.

X = μT + Bias + Precision

⇔ X − μT = Bias + Precision

⇔ X − μT = Total error

⇔ X − μT = Measurement Error

4.5.4 Computational AspectsAs described in Section 4.1, the classical design of a pre-study validation consists inperforming different runs with replicates in each run. Let’s note p the number of runs, ni

the number of replicates in the ith run, N = n1 + · · · + np the total number ofmeasurements, and xij the jth measurement in the ith run.

Validation criteria such as accuracy and precision are estimated for each concentrationlevel by statistical analysis of the back-calculated quantities. Computationally, a one-factorrandom effects Analysis of Variance (ANOVA) model is fit to the back-calculated values ateach level with run as the random effects factor:

xij = μ + αi + εij ,

where μ is the mean of calculated concentrations, αi and εij are normally distributed withmean 0 and variances σ2

B and σ2W , respectively. Here σ2

B is the run-to-run variance and σ2W

is the within-run variance. The estimates of μ, σ2B, and σ2

W are given by:

μ = x·· =1N

p∑i=1

nixi·, σ2W =

1N − p

p∑i=1

ni∑j=1

(xij − xi·)2,

σ2B =

p − 1N − n

[(1

p − 1

p∑i=1

(xi· − x··)2

)− σ2

W

],

where xi· = n−1i

∑ni

j=1 xij and n = N−1 ∑pi=1 ni. By definition, the estimate of the within-run

variance corresponds to the variance of repeatability and the sum of the within-run andrun-to-run components corresponds to the intermediate precision. That is, the intermediateprecision variance is σ2

B + σ2W .

The relative error (RE), the coefficient of variation of the intermediate precision (CVIP ),and the total error (TE) are calculated as follows:

RE = 100μ − μ

μ, CVIP = 100

√σ2

W + σ2B

μ, TE = RE + CVIP .

Program 4.7 utilizes these formulas to compute the accuracy, precision, and total errorperformance parameters. In this example, each combination of series and plate is consideredas a run. Again, this program uses the CALC CONC data set created in Program 4.5.

Page 100: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 87

Program 4.7 Computation of the accuracy and precision parameters

proc sort data=calc_conc;by concentration;

proc mixed data=calc_conc;class run;model calc_conc=/solution;random run;by concentration;ods output dimensions=dim;ods output solutionf=solf;ods output CovParms=var_comp;

proc sort data=calc_conc;by concentration run plate;

proc univariate data=calc_conc noprint;var calc_conc;by concentration run;output out=stat_by_conc_run n=n;

proc sql;create table stat_by_conc asselect t1.concentration, cap_n, sum_n_sq/cap_n as n, n_run, mean,

sqrt(sigma_w_sq) as sigma_w,100*sqrt(sigma_w_sq)/t1.concentration as cv_w,sqrt(sigma_b_sq) as sigma_b,100*sqrt(sigma_b_sq)/t1.concentration as cv_b,sqrt(sigma_w_sq+sigma_w_sq) as sigma_t,100*sqrt(sigma_w_sq+sigma_w_sq)/t1.concentration as cv_t

from (select concentration, value as cap_n from dim wheredescr=’Observations Used’) as t1,(select concentration, estimate as mean from solf) as t2,(select concentration, sum(n**2) assum_n_sq from stat_by_conc_run group by concentration) as t3,(select concentration, estimate as sigma_b_sq from var_compwhere covparm=’run’) as t4,(select concentration, estimate as sigma_w_sq from var_compwhere covparm=’Residual’) as t5,(select count(run) as n_run from (select distinct run fromstat_by_conc_run)) as t6 where t1.concentration=t2.concentration=t3.concentration=t4.concentration=t5.concentration;

run;quit;

* Calculation of validation criteria;data stat_by_conc;

set stat_by_conc;format mean re cv_w cv_b cv_t te 4.1 low_tol_lim_rel upp_tol_lim_rel 5.1;re=100*(mean-concentration)/concentration;te=abs(re)+cv_t;r=(sigma_b/sigma_w)**2;b=sqrt((r+1)/(n*r+1));ip=sigma_b**2+sigma_w**2;ddl=((r+1)**2)/(((r+1/n)**2)/(n_run-1)+(1-1/n)/(cap_n));low_tol_lim_abs=mean-tinv(0.975,ddl)*sqrt(ip*(1+1/(cap_n*b**2)));upp_tol_lim_abs=mean+tinv(0.975,ddl)*sqrt(ip*(1+1/(cap_n*b**2)));low_tol_lim_rel=(low_tol_lim_abs-concentration)*100/concentration;upp_tol_lim_rel=(upp_tol_lim_abs-concentration)*100/concentration;

Page 101: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

88 Pharmaceutical Statistics Using SAS: A Practical Guide

label concentration=’Nominal*concentration’mean=’Calculated*concentration’re=’Relative*error (%)’cv_w=’Within-run*CV (%)’cv_b=’Run-to-run*CV (%)’cv_t=’Intermediate*precision*CV (%)’te=’Total*error(%)’low_tol_lim_rel=’Upper 95%*tolerance*limit’upp_tol_lim_rel=’95% TI*upper*limit’;

proc print data=stat_by_conc noobs split=’*’;format mean re cv_w cv_b cv_t te 4.1 low_tol_lim_rel upp_tol_lim_rel 5.1;var concentration mean re cv_w cv_b cv_t te;run;

Output from Program 4.7

IntermediateNominal Calculated Relative Within-run Run-to-run precision Total

concentration concentration error (%) CV (%) CV (%) CV (%) error(%)

3.5 3.2 -7.3 37.7 23.0 53.3 60.67.0 8.2 16.6 14.7 18.5 20.8 37.5

14.1 15.5 9.8 12.7 8.8 18.0 27.828.0 30.0 7.3 4.3 5.2 6.1 13.456.0 57.9 3.3 2.7 5.3 3.9 7.2113.0 114 1.0 2.4 6.2 3.4 4.3225.0 235 4.6 3.7 4.5 5.2 9.8450.0 469 4.1 5.5 5.0 7.7 11.9

Output 4.7 shows the estimates of the main accuracy, precision, and total errorperformance parameters of the method as a function of the estimated concentration. It isclear that, at low concentration levels (less than or equal to 14.1 μM), either the truenessor precision is not acceptable. This is well summarized by the total error that becomessmaller than 30% for concentration values greater than 14.1 μM. This already gives an ideaabout the capability of the method, but as it will be shown in the following section, thedecision rule should be established on a more refined method that takes the notion of riskinto consideration.

4.6 Decision RuleIt was mentioned in the Introduction that Equation (4.2), which describes the mainobjective of an analytical method, cannot be solved exactly. A simple way to resolve thisproblem and make a reliable decision, proposed by several authors (Hubert et al., 2004;Boulanger et al., 2000a, 2000b; Hoffman and Kringle, 2005), relies on computing theβ-expectation tolerance intervals (Mee, 1984):

EμM ,σM(PX [μM − kσM < X < μM + kσM |μM , σM ]) = β,

where the k factor is determined so that the expected proportion of the population fallingwithin the interval is equal to β. If the β-expectation tolerance interval is totally includedwithin the acceptance limits [−λ,+λ], i.e., if μM − kσM > −λ and μM + kσM < λ, theexpected proportion of measurements within the same acceptance limits is greater than orequal to β. Note that the opposite statement is not true, i.e., if either μM − kσM < −λ orμM + kσM > λ, the expected proportion is not necessarily smaller than β.

Most of the time, an analytical procedure is intended to quantify over a range ofquantities or concentrations. Consequently, during the validation phase, samples are

Page 102: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 89

prepared to adequately cover this range, and a β-expectation tolerance interval iscalculated at each level.

A measurement error profile is obtained, on one hand, by connecting the lower limitsand, on the other hand, by connecting the upper limits. A procedure is valid over a certainrange of values if the measurement error profile is included within the acceptance limits[−λ,+λ]. This concept is illustrated in Figure 4.5 for a chromatographic bio-analyticalmethod (Hubert et al., 1999). In the left panel of Figure 4.5, the measurement error profileis included within the 20% acceptance limits over the entire range of concentrations, andthus the method is valid to quantify concentration values over this range. However, in theright panel of Figure 4.5, the method can accurately quantify only concentration valuesgreater than 300μM.

Figure 4.5 Measurement error profiles with observations, 20% acceptance limits (dotted lines), β-expectationtolerance limits (solid curves), and biases (dashed curves) expressed in terms of relative errors as a function ofthe concentration values for a chromatographic bio-analytical procedure

Rel

ativ

e E

rror

(%

)

-40

-20

0

20

40

Concentration

0 200 400 600 800 1000

Rel

ativ

e E

rror

(%

)

-40

-20

0

20

40

Concentration

0 200 400 600 800 1000

A measurement error profile gives the analyst a sense of what a procedure will be ableto produce over the intended range. The interpretation of a measurement error profile isthat it shows where 100β% of the measures provided by this analytical method will lie,which is directly connected to the objective of the analytical method (produce measuresclose to the unknown true values).

In practice, the β-expectation tolerance interval is obtained as follows (on a relativescale):[

RE − Qt

(v,

1 + β

2

) √1 +

1pnB2 CVIP ,RE + Qt

(v,

1 + β

2

) √1 +

1pnB2 CVIP

],

where p is the number of runs, n is the number of replicates within each run, Qt(a, b) is the100b% quantile of the t distribution with a degrees of freedom, and

R =σ2

B

σ2W

, B =√

R + 1nR + 1

, v = (R + 1)2[(R + 1/n)2

p − 1+

1 − 1/n

pn

]−1

.

Program 4.8 lists and plots the β-expectation tolerance limits for this study consideredin Program 4.7 (the limits are included in the STAT BY CONC data set created byProgram 4.7).

Page 103: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

90 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 4.8 Computation of the β-expectation tolerance limits

proc print data=stat_by_conc noobs split=’*’;var concentration mean low_tol_lim_rel upp_tol_lim_rel;run;

axis1 minor=none label=(angle=90 "Total error (%)") order=(0 to 70 by 10);axis2 minor=none label=("Nominal concentration") logbase=10 logstyle=expand;symbol1 value=dot i=join color=black line=1 width=3 height=3;proc gplot data=stat_by_conc;

format te 4.0;plot te*concentration/frame haxis=axis2 vaxis=axis1 vref=30 lvref=34;run;quit;

axis1 minor=none label=(angle=90 "Relative error (%)") order=(-120 to 100 by 20);axis2 minor=none label=("Nominal concentration") logbase=10 logstyle=expand;symbol1 value=dot i=join color=black line=1 width=3 height=3;symbol2 value=none i=join color=black line=8 width=3;symbol3 value=dot i=join color=black line=1 width=3 height=3;proc gplot data=stat_by_conc;

format low_tol_lim_rel re upp_tol_lim_rel 4.0;plot (low_tol_lim_rel re upp_tol_lim_rel)*concentration/overlay frame

haxis=axis2 vaxis=axis1 vref=-30,30 lvref=34;run;quit;

Output from Program 4.8

Lower 95% Upper 95%Nominal Calculated tolerance tolerance

concentration concentration limit limit

3.5 3.2 -103 88.47.0 8.2 -37.1 70.4

14.1 15.5 -23.8 43.428.0 30.0 -8.1 22.756.0 57.9 -10.8 17.4

113.0 114 -15.0 16.9225.0 235 -8.5 17.7450.0 469 -12.2 20.5

Output 4.8 shows that the tolerance interval is fully included within the 30% acceptancelimits for concentration levels strictly greater than 14.1 μM. It is also important to noticethat once the tolerance interval is fully included within the acceptance limits, the classicalvalidation criteria are also satisfied. That is, the accuracy and intermediate precision arebetter than the usually recommended 15% threshold. Figure 4.6 provides a graphicalsummary of the quantities produced by Program 4.8. In the right panel of Figure 4.6, thetwo horizontal (dotted) lines represent the 30% acceptance limits and are equivalent to theacceptance limits in Figure 4.4 on an absolute scale. The dashed curve represents theaccuracy (relative error).

Program 4.9 computes the individual total and individual relative errors and plots themalong with the total error and measurement error curves displayed in Figure 4.6. Theresulting total error and measurement error profiles are shown in Figure 4.7.

Page 104: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 91

Figure 4.6 Total error (left panel) and measurement error (right panel) profiles

Tot

al e

rror

(%

)

0

10

20

30

40

50

60

70

Nominal concentration

1 10 100 1000

Rel

ativ

e er

ror

(%)

-120

-100

-80

-60

-40

-20

0

20

40

60

80

100

Nominal concentration

1 10 100 1000

Program 4.9 Computation of individual total and individual relative errors

proc sql;create table te asselect distinct 2 as type, calc_conc.concentration,

abs(calc_conc-calc_conc.concentration)*100/calc_conc.concentration as tefrom calc_conc, stat_by_concwhere calc_conc.concentration=stat_by_conc.concentration

unionselect 1 as type, concentration, te from stat_by_conc;run;

quit;axis1 minor=none label=(angle=90 "Total error (%)") order=(0 to 100 by 20);axis2 minor=none label=("Nominal concentration") logbase=10 logstyle=expand;symbol1 value=dot i=join color=black line=1 width=3;

symbol2 value=circle i=none color=black height=5;proc gplot data=te;

plot te*concentration=type/vaxis=axis1 haxis=axis2 vref=30 lvref=34 nolegend;

run;quit;

data me;

set stat_by_conc;array y(3) low_tol_lim_rel re upp_tol_lim_rel;do j=1 to 3;

me=y(j); type=j;

output;end;

proc sql;

create table me asselect distinct 4 as type, calc_conc.concentration,

(calc_conc-calc_conc.concentration)*100/calc_conc.concentration as me

from calc_conc, stat_by_concwhere calc_conc.concentration=stat_by_conc.concentrationunion

select type, concentration, me from me;run;quit;

Page 105: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

92 Pharmaceutical Statistics Using SAS: A Practical Guide

axis1 minor=none label=(angle=90 "Relative error (%)") order=(-120 to 100 by 20);

axis2 minor=none label=("Nominal concentration") logbase=10 logstyle=expand;symbol1 value=dot i=join color=black line=1 width=3 height=5;symbol2 value=none i=join color=black line=20 width=3;

symbol3 value=dot i=join color=black line=1 width=3 height=5;symbol4 value=circle i=none color=black height=5;proc gplot data=me;

plot me*concentration=type/frame haxis=axis2 vaxis=axis1 nolegend vref=-30,30 lvref=34;

run;quit;

Figure 4.7 demonstrates that most of the individual relative errors are within thetolerance limits. This highlights the fact that both the total error and measurement errorprofiles provide clues about how extreme results may be when produced by an analyticalmethod. That is, most results will be within the calculated limits. The measurement errorprofile is preferred because it is predictive (with a specified risk) of future results obtainedby the method, while the total error profile is more descriptive.

Figure 4.7 Total error (left panel) and measurement error (right panel) profiles with individual total andindividual relative errors

Tot

al e

rror

(%

)

0

20

40

60

80

100

Nominal concentration

1 10 100 1000

Rel

ativ

e er

ror

(%)

-120

-100

-80

-60

-40

-20

0

20

40

60

80

100

Nominal concentration

1 10 100 1000

The choice of the criteria depends on the analyst’s preference. If a certain amount ofrisk is associated with the decision, the measurement error profile should be used.Otherwise, the total error profile could be considered.

In conclusion, the measurement error profile is an easy-to-interpret tool that allows theanalyst to check trueness, precision, and accuracy of the method. Moreover, it isstatistically meaningful and objectively oriented. Note that the lower and upper limits ofquantitation can be estimated using either total error and measurement error profiles byintersecting the profiles with the acceptance limits.

4.7 Limits of Quantification and Range of the AssayThe upper and lower limits of quantitation (ULOQ and LLOQ) of an analytical procedureare the lowest and highest amounts of the targeted substance in the sample that can bequantitatively determined under the prescribed experimental conditions. As a consequence,the range of an analytical procedure is the range between the lower and upper limits ofquantitation for which the analytical procedure was demonstrated to have a suitable levelof measurement error.

Page 106: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 93

In practice, the information needed to establish the limits of quantitation and theassociated range is already available in the measurement profile plot. As proposed byHubert et al. (1999) and Hubert et al. (2004), the limits of quantitation are the mostextreme (low, high) concentrations (quantities) at which the tolerance interval is stillwithin the acceptance limits, should the tolerance limits cross the acceptance limits. If alltolerance limits lie within the acceptance limits, the limits of quantitation are defined asthe most extreme quantities tested in the study.

Figure 4.8 Derivation of the limits of quantification and range of the assay using the measurement error profile

LLOQ ULOQ

Rel

ativ

e er

ror

(%)

-120

-100

-80

-60

-40

-20

0

20

40

60

80

100

Nominal concentration

1 10 100 1000

Figure 4.8 shows how the upper and lower limits of quantitation are defined. In thisexample, the lower limit is approximately 24 μM, and the upper limit is the maximumconcentration investigated in this study, i.e., 450 μM.

4.8 Limit of DetectionThe limit of detection (LOD) of an analytical procedure is the lowest amount of thetargeted substance in the sample that can be detected reliably, but not necessarilyquantified as an accurate value using the experimental conditions prescribed. A variety ofmethods to estimate the LOD have been proposed in the literature, generally based on thecalibration information and estimates. None of them are really satisfactory and aresensitive to various assumptions related to the model, the design of experiment andmodeling of heterogeneity of variances. Based on our experience, the “best” and mostconsistent estimate proposed for bioanalytical methods aiming at covering a large range ofconcentrations is based on dividing the lower limit of quantitation by 3 or 3.33. This isjustified by the fact that it is largely accepted that the LOD is three times the noise of thesignal and the LLOQ is ten times the same noise.

So, for the example used in Program 4.8, the LOD can be established around 8 μM.This is consistent with Figure 4.2. The 8 μM level appears to be the lowest concentrationlevel producing a signal that is clearly above the signals in the lower asymptote.

4.9 SummaryThroughout this chapter we have proposed ways to understand, estimate, and interpretvarious criteria required for assessing the validity of an analytical method. These criteriamust be reported, and the code used to compute the criteria must be documented

Page 107: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

94 Pharmaceutical Statistics Using SAS: A Practical Guide

according to best practices. Regardless of the complexity of computations and models, theobjective of an analytical method (are the measurement errors acceptable?) should neverbe forgotten and should remain the primary focus. The information needed to make adecision is contained in the measurement error profile. All performance criteria—linearity,accuracy, precision, limits, measurement errors—can be assessed using a graphical profileand can be easily understood and interpreted by an analyst.

Another important remark related to the decision is that, if the tolerance intervals arewithin the acceptance limits, all of the required criteria are guaranteed to be met. Theopposite, however, is not true. That is, even when all of the performance criteria aresatisfied, the measurement errors will not necessarily be acceptable. Although it is commonto assume that good methods will always produce good results and most regulatorydocuments were written in this spirit, it is important to remember that only the oppositestatement holds true: good results can only be obtained with a good method.

4.10 TerminologyAnalytical procedure (or method or assay). Written procedure which describes all themeans and the operating procedures required to perform the analysis of the analyte. Thatis, field of application, principle and/or reactions, definitions, reactants, equipments,operating procedures, expression of results, suitability tests, test reports.Analyte or activity. The analyte (or activity when relevant) is the matter of theanalytical procedure. The analyte is a physical entity (e.g., water activity), chemical entity(e.g., active substance alone or in a pharmaceutical formulation, total lipids, aspartame,lead) or biological entity (e.g., atpmetric activity). In the case of quantitative analyticalprocedures of farm and food products, “analyte” is equivalent to “measurand”.Matrix. All constituents of the laboratory sample other than the analyte. A type of matrixis defined as a group of materials or of products considered by the analyst as having ahomogeneous behavior with regard to the analytical method used.Blank. Test performed in the absence of the matrix (blank reactant) or in a matrixwithout analyte (blank matrix). By extension, the instrument response in the absence ofthe analyte is used (blank instrumental).Accepted reference value. An accepted reference value is a value used as a reference,agreed for a comparison and derived from:

• A theoretical or established value, based on scientific reasons.• An assigned or certified value, based on experimental data from a national or

international organization.• A consensus value, based on a collaborative experimental work.• The mathematical expectation of the (measurable) quantity, in cases where the previous

points are not applicable, i.e., the mean of a specified population of determinations (cf.NF ISO 5725-1).

Calibration standard (or calibration sample). Calibration standards are samples ofknown concentrations, with or without matrix, that allow drawing the calibration curve.Validation standard (or validation sample). Validation standards are samplesreconstituted in the matrix or in any other reference material with true values set byconsensus and used to validate the analytical procedure.

ReferencesBelanger, B.A., Davidian, M., Giltinan, D.M. (1996). “The effect of variance function estimation on

nonlinear calibration inference in immunoassay data.” Biometrics. 52, 158–175.

Page 108: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 4 Statistical Considerations in Analytical Method Validation 95

Boulanger, B., Hubert, P., Chiap, P., Dewe, W. (2000a). “Objectives of pre-study validation anddecision rules.” Presented at AAPS APQ Open forum, Indianapolis, Indiana.

Boulanger, B., Hubert, P., Chiap, P., Dewe, W., Crommen, J. (2000b). “Analyse statistique desresultats de validation de methodes chromatographiques.” Presented at Journees du GMP,Bordeaux, France.

DeSilva, B., Smith, W., Weiner, R., Kelley, M., Smolec, J., Lee, B., Khan, M., Tacey, R., Hill, H.,Celniker, A. (2003). “Recommendations for bioanalytical method validation of ligand-bindingassays to support pharmacokinetic assessments of macromolecules.” Pharmaceutical Research.20, 1885–1900.

FDA (Food and Drug Administration). (2001). Guidance for industry: Bioanalytical methodsvalidation.

Finney, D. (1978). Statistical Methods in Biological Assays. London: Charles Griffin.Findlay, J.W.A., Smith, W.C., Lee, J.W., Nordblom, G.D., Das, I., Desilva, B.S., Khan, M.N.,

Bowsher, R.R. (2001). “Validation of immunoassays for bioanalysis: a pharmaceutical industryperspective.” Journal of Pharmaceutical and Biomedical Analysis. 21, 1249–1273.

Hoffman, D., Kringle, R. (2005). “Two-sided tolerance intervals for balanced and unbalancedrandom effects models.” Journal of Biopharmaceutical Statistics. 15, 283–293.

Hubert, P., Chiap, P., Crommen, J., Boulanger, B., Chapuzet, E., Mercier, N., Bervoas-Martin, S.,Chevalier, P., Grandjean, D., Lagorce, P., Lallier, M., Laparra, M.C., Laurentie, M., Nivet, C.(1999). “The SFSTP guide on the validation of chromatographic methods for drug bioanalysis:from the Washington Conference to the laboratory.” Analytica Chimica Acta. 391, 135–148.

Hubert, P. Nguyen-Huu, J.-J., Boulanger, B., Chapuzet, E., Chiap, P., Cohen, N., Compagnon,P.-A., Dewe, W., Feinberg, M., Lallier, M., Laurentie, M., Mercier, N., Muzard, G., Nivert, C.,Valat, L. (2004). “Harmonization of strategies for the validation of quantitative analyticalprocedures. An SFSTP proposal Part I.” Journal of Pharmaceutical and Biomedical Analysis.36, 579–586.

ICH (International Conference on Harmonization). (1995). Guideline on validation of analyticalprocedures: definitions and terminology.

ICH (International Conference on Harmonization). (1997). Guideline on validation of analyticalprocedures: methodology.

International Organization for Standardization. ISO 5725-1:1994 Accuracy (Trueness and Precision)of Measurement Methods and Results—Part 2: Basic Method for the Determination ofRepeatability and Reproducibility of a Standard Measurement Method [Edition 1].

Lee, J.W., Nordblom, G.D., Smith, W.C., Bowsher, R.R. (2003). “Validation of bioanalytical assaysfor novel biomarkers: Practical recommendations for clinical investigation of new drug entities.”In Biomarkers in Clinical Drug Development. Bloom, J., Dean, R.A., eds. New York: MarcelDekker, 119–148.

Mee, R.W. (1984). “β-expectation and β-content tolerance limits for balanced one-way ANOVApandom model.” Technometrics. 26, 251–254.

Mire-Sluis, A.R, Barrett, Y.C., Devanarayan, V., Koren, E., Liu, H., Maia, M., Parish, T., Scott,G., Shankar, G., Shores, E., Swanson, S.J., Taniguchi, G., Wierda, D., Zuckerman, L.A. (2004).“Recommendations for the design of immunoassays used in the detection of antibodies againstbiological products.” Journal of Immunological Methods. 289, 1–16.

O’Connell, M.A., Belanger, B.A., Haaland P.D. (1993). “Calibration and assay development usingthe four-parameter logistic model.” Chemometrics and Intelligent Laboratory Systems. 20,97–114.

SAS Institute Inc. SAS/STAT User’s Guide. Cary, NC: SAS Institute Inc.Smith, W.C., Sittampalam, S.G. (1998). “Conceptual and statistical issues in the validation of

analytical dilution assays for pharmaceutical applications.” Journal of BiopharmaceuticalStatistics. 8, 509–532.

Page 109: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 110: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5

Some StatisticalConsiderations in NonclinicalSafety Assessment

Wherly HoffmanCindy LeeAlan ChiangKevin GuoDaniel Ness

5.1 Overview of Nonclinical Safety Assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97

5.2 Key Statistical Aspects of Toxicology Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98

5.3 Randomization in Toxicology Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99

5.4 Power Evaluation in a Two-Factor Model for QT Interval . . . . . . . . . . . . . . . . . . . . . . . . . . 102

5.5 Statistical Analysis of a One-Factor Design with Repeated Measures . . . . . . . . . . . . . . . 106

5.6 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

Understanding the intrinsic toxicological properties of chemicals is fundamental toevaluating their safety. Such data are derived from toxicological studies that are requiredby national and international regulatory organizations. These data are used to determinethe potential hazards and to gain an understanding of the potential risks to humans. Thischapter provides a brief overview of the role of nonclinical safety assessment in drugdevelopment and covers several important statistical considerations in defining hazards.The topics include randomization, power evaluation, and data analysis. Examples are giventhroughout the chapter to illustrate the described statistical methods for each topic.

5.1 Overview of Nonclinical Safety AssessmentThe goal of drug development is to identify safe and efficacious treatments for humandiseases. Safety and efficacy are assessed through careful evaluation in animal models andin vitro systems (nonclinical setting) and in exposed human populations (clinical setting)

Wherly Hoffman is Head, Toxicology/Drug Disposition and Animal Health Statistics, Eli Lilly, USA. Cindy Lee is AssociateSenior Statistical Analyst, Toxicology Statistics, Eli Lilly, USA. Alan Chiang is Senior Research Scientist, ToxicologyStatistics, Eli Lilly, USA. Kevin Guo is Assistant Senior Statistician, Toxicology Statistics, Eli Lilly, USA. Daniel Ness isResearch Advisor, Toxicology, Eli Lilly, USA.

Page 111: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

98 Pharmaceutical Statistics Using SAS: A Practical Guide

throughout drug development and post-marketing. This chapter focuses on the design andanalysis of studies that are integral to nonclinical safety assessment.

The objectives in nonclinical safety assessment are to define the toxicity profile ofcandidate drugs, estimate the margin of safety by understanding the relationship betweentoxic exposures and efficacious exposures, and provide a judgment on the likelihood thatthe animal findings can be extrapolated to humans. These assessments are important

• prior to candidate selection because they provide internal decision-makers with aprobability of technical success for the new compound,

• during clinical development because they protect the safety of individuals in clinicaltrials, and

• during the post-marketing phase because they support alternative formulations orclinical indications or further investigation of newly discovered safety issues.

It is beyond the scope of this chapter to consider all questions asked in nonclinicalstudies; however, a few questions are particularly central to safety assessment. Theseinclude:

• What dose or plasma exposure of the experimental drug is not associated with anyadverse outcome (i.e., the no observed adverse effect level, or NOAEL)?

• What dose or plasma exposure of drug does not result in any observed biological effect(i.e., the no observed effect level, or NOEL)?

• What is the maximum tolerated dose (MTD)?• What is the nature of the dose-response relationship? For example, how steep is the

dose-response curve as judged by the difference between the NOAEL and the MTD, oris the response monotonic or nonmonotonic?

• What are the target organs of toxicity?• Are the toxicities monitorable? That is, are there antemortem measures that are

predictive of tissue pathology?• Are the toxicities reversible?

Most of the studies designed to answer the questions above are mandated by regulatoryagencies worldwide. Guidance documents for pharmacology and toxicology are availableelectronically via the Food and Drug Administration (FDA), European Medicines Agency(EMEA), and the International Conference on Harmonization (ICH) Web sites. Theseguidance documents assist in determining the relevant single-dose, repeat-dose, genetictoxicity, safety pharmacology (e.g., central nervous system, cardiovascular, and respiratory)studies, immunotoxicity, reproductive/developmental, and carcinogenicity studies as well asthe necessary studies to ensure adequate quality of the formulated material (e.g., impurityqualification). Other studies to examine more closely certain mechanisms of toxicity orcompound-specific issues are conducted as needed.

5.2 Key Statistical Aspects of Toxicology StudiesThe key statistical aspects of the design and analysis of toxicology studies to be discussedare randomization, power evaluation, and data analysis.

5.2.1 RandomizationSince proper randomization is the foundation of valid statistical inference, this chapterstarts with the randomization methods for two commonly used designs in toxicology,namely, the parallel design and Latin square design.

Page 112: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 99

Parallel designs include studies with one factor, two factors, one-factor with repeatedmeasurements, and more. In a parallel design, separate groups of animals are assigned tocombinations of factor levels. For example, in a one-factor design with four dose groups,there would be four different groups of animals, each receiving one of the four doses. For atwo-factor design with four dose groups and two routes of delivery, there would be eightdifferent groups of animals, each receiving one of the eight combinations of the four dosesand two routes.

Latin square designs are efficient for incorporating three factors each with the samenumber of levels. For example, a basic 4 × 4 Latin square design can accommodate fouranimals, each receiving one of four treatments in four dosing periods. This design allows forthe evaluation of the treatment, animal, and time effects. However, one has to assume thatall two- or three-way interactions are not significant.

The random assignment of animals to the groups for the parallel and Latin squaredesigns is discussed in Section 5.3.

5.2.2 Power EvaluationPower evaluation characterizes the strength of an inferential test. It is a function of the sizeof the change to be detected, the variability, the Type I error rate, and the sample size.Although certain sample sizes in standard toxicology studies are commonly accepted andbased, in part, on regulatory guidance, many toxicology studies targeted at specialendpoints merit an assessment of the power and sample size. An example of this is theevaluation of QT prolongation in large animal toxicology studies with four treatmentgroups and three or four animals per group for each sex. The QT interval is a measure ofthe time between the start of the Q wave and the end of the T wave in the heart’selectrical cycle. The details of the study design and statistical tests are described inSection 5.4. The evaluation of power is performed in a two-factor analysis of variance(ANOVA) framework by simulation.

5.2.3 Data AnalysisFor each well-designed study, the statistical hypotheses and tests are defined in theprotocol. The most commonly collected data for toxicology studies is body weight. A bodyweight change, either a gain or loss, is important for monitoring the well-being of theanimal and toxicities of a compound. Section 5.5 describes statistical methods used in theanalysis of body weight data in toxicology studies based on a one-factor ANOVA modelwith repeated measures.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

5.3 Randomization in Toxicology StudiesOne of the most important elements of any research design is the concept ofrandomization. This concept is central to all types of toxicology research (Lin, 2001).Random assignment of experimental animals to different groups helps reduce the potentialbiases among the comparison groups and allows a valid interpretation of the researchresults. Most statistical packages can produce random numbers within a specified range,which can be used to assign experimental units to treatments. Some textbooks have tablesof random numbers designed for this purpose.

There are generally two approaches to creating randomization tables in SAS. The firstapproach is based on a direct generation of randomization tables using a DATA step. Theother approach relies on the PLAN procedure that provides different randomization layouts

Page 113: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

100 Pharmaceutical Statistics Using SAS: A Practical Guide

depending on the experiment. For a more detailed description of random allocationmethods in pre-clinical and clinical studies, see Chapter 9, “Allocation in RandomizedClinical Trials”.

5.3.1 Block Randomization in a Parallel DesignIn this subsection, the randomized complete block design is discussed to illustrate therandomization approach. Consider designing a study to determine whether different doselevels of a compound affect the liver weight of a mouse after one week of treatment. If fourdose levels, including a control, are evaluated and 48 mice are available for the study, thenthese mice would need to be assigned to the four dose groups. Since the liver weight isrelated to the body weight, a common practice is to assign the animals to treatments usinga randomized complete block design with body weight stratification.

A randomized complete block design structure is any blocking scheme in which thenumber of experimental units within a block is a multiple of the number of treatments, andthus the experimental units can be assigned completely at random to a complete set oftreatments in each block.

Program 5.1 creates a randomization table for the study using a DATA step. First, theprogram defines the allocation number for each animal based on the animal id and sortsthe animals by their body weights. The random variable RAND is created using theRANUNI function that generates a random number from the uniform distribution on the(0, 1) interval. The BLOCK variable assigns each animal to a block: the first four mice toBlock 1 (the four lightest animals), the second four mice to Block 2, etc. The RANKprocedure ranks the random numbers from the smallest to the largest within each block.The ranking of the random numbers within a block is used for treatment assignment.

Program 5.1 Randomization using a DATA step

data bodyweight;animal_id=_n_;input bw @@;datalines;

22.1 25.5 24.2 26.1 23.3 22.0 21.8 24.8 23.1 23.0 23.0 24.825.3 25.6 24.8 24.5 26.0 23.6 26.3 24.0 26.9 25.9 22.7 22.422.5 22.3 22.3 25.5 20.9 24.5 22.2 23.3 20.3 26.3 27.6 26.526.8 25.6 26.6 23.5 22.4 21.3 23.7 26.8 24.6 24.2 26.1 26.2;proc sort data=bodyweight;

by bw;data bdwt;

set bodyweight;rand=ranuni(1202019);block=1+int((_n_-1)/4);

proc rank data=bdwt out=bwstrat (drop=rand);by block;var rand;ranks group;

proc sort data=bwstrat;by group animal_id;

proc print data=bwstrat noobs label;label animal_id="Animal ID"

bw="Body weight"block="Block"group="Treatment group";

run;

Page 114: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 101

Partial output from Program 5.1

Animal Body TreatmentID weight Block group

10 23.0 4 112 24.8 8 118 23.6 5 120 24.0 6 124 22.4 3 128 25.5 9 129 20.9 1 131 22.2 2 137 26.8 12 139 26.6 11 145 24.6 7 148 26.2 10 12 25.5 8 27 21.8 1 2

17 26.0 10 2

Output 5.1 displays the first 15 rows in the randomization table generated byProgram 5.1.

5.3.2 Randomization in a Latin Square DesignThe other popular design used in toxicology studies is the Latin square design. The Latinsquare design consists of blocking in two directions. For an experiment involving ntreatments, n2 experimental units are assigned into an n × n square in which the rows arecalled row blocks and the columns are called column blocks. Thus, the n × n arrangementof experimental units is blocked in two directions. In a Latin square design, the treatmentsare randomly assigned to animals in the square such that each treatment occurs once andonly once in each row block and once and only once in each column block. See Box et al.(1978) for various arrangements of treatments into row and column blocks.

The %LATINSQ macro in Program 5.2 uses PROC PLAN to generate an n × n Latinsquare design. The macro includes two arguments: N is the dimension of the Latin square,and SEED is the seed for randomization.

The FACTORS statement in PROC PLAN specifies the row and column blocks of thedesign (ANIMAL ID and PERIOD). The TREATMENTS statement specifies thetreatment groups (GROUP). The OUTPUT statement saves the design generated to thespecified data set (LATIN). Creating a randomization schedule for a Latin square designinvolves randomly permuting the row, column, and treatment values independently. Toaccomplish this, the association type of each factor is specified as RANDOM in theOUTPUT statement. The output is summarized using the TABULATE procedure.

Page 115: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

102 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 5.2 Randomization in an n × n Latin square design

%macro latinsq(n,seed);proc plan seed=&seed;

factors animal_id=&n ordered period=&n ordered/noprint;treatments group=&n cyclic;output out=latin period random animal_id random group random;

proc tabulate data=latin formchar=’ ’;label animal_id="Animal ID" period="Period";keylabel sum=’ ’;class period animal_id;var group;table animal_id, period*(group=’’*f=3.)/rts=8;run;

%mend latinsq;%latinsq(n=4,seed=1034567);

Output from Program 5.2

Period

1 2 3 4

Animal ID

1 1 3 4 22 3 4 2 13 2 1 3 44 4 2 1 3

5.4 Power Evaluation in a Two-Factor Model for QT IntervalThis section describes the process of estimating the power of a trend test in a two-factorANOVA model. The application is for a general toxicology study in beagle dogs with avehicle control (Dose 0) and three groups of increasing doses of a compound (Doses 1, 2,and 3). A sample size of three or four animals per sex per group is generally used. Thepurpose of the analysis is to evaluate the treatment effects on heart rate-corrected QTintervals by identifying the highest no observed effect dose level (NOEL). The powerevaluation is important because the recent ICH S7B guideline (2004) recommends that thesensitivity and reproducibility of the in vivo test system be characterized. See also Chianget al. (2004).

The QT interval of the electrocardiogram (ECG) is a common end-point forcharacterizing potential drug-associated delayed ventricular repolarization in vivo (ICHS7B, 2004). It has been conjectured that delayed ventricular repolarization caused by acompound may lead to serious ventricular tachyarrhythmias in humans (see, for example,Kinter, Siegl and Bass, 2004). Statistical analysis of QT interval data is complicated by thefact that the analysis is inversely correlated with heart rate (HR). Therefore, analysis ofQT interval data generally includes an adjustment for the RR interval (the RR interval,expressed in seconds, is equal to 60 times the reciprocal of heart rate, i.e., RR=60/HR).Fridericia’s formula (QTc = QT/ 3

√RR, Fridericia, 1920) is commonly used to obtain the

heart rate-corrected QT intervals in nonclinical evaluation.

Page 116: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 103

5.4.1 Sequential Testing MethodAt Eli Lilly and Company, QT intervals in a general toxicology study are collected atpre-specified time points both before and after treatment on selected dosing dates. QTcdata at each time point are analyzed using a two-factor ANOVA model. Factors in themodel include treatment, sex, and the interaction of those two factors. Effects associatedwith treatment and treatment-by-sex interaction are tested using an F -test at the 0.05significance level. Monotonicity of dose response is examined by first testing for aninteraction between the treatment linear trend and sex group at the 0.05 significance level.If this interaction is significant, a sequential trend test (Tukey et al., 1985) on treatmentmeans is performed at the 0.05 significance level for each sex group and for the two sexgroups combined. Otherwise, the sequential trend test is performed only for the combinedgroup. For a detailed description of linear and other trend tests in dose-ranging studies, seeChapter 11, “Design and Analysis of Dose-Ranging Clinical Studies”.

To define the sequential testing procedure in the two-factor ANOVA model, consider thefollowing four tests:

Test A. An interaction between the treatment linear trend and sex.Test B. The overall treatment linear trend for combined sexes.Test C. The treatment linear trend for each sex.Test D. Test C when Test A is significant or Test B when Test A is not significant.

Figure 5.1 Flow chart for analysis of two-factor ANOVA

Linear trend by sexinteraction test

(Test A)

Significant at 0.05?

Yes

No

�Linear trend test

for each sex(Test C)

Linear trend testfor combined sexes

(Test B)

Figure 5.1 is a flow chart of the sequential testing procedure. Test D represents anoverall assessment of the study and is of primary interest.

The sequential trend test by Tukey et al. (1985) is carried out as follows. Let μ0 denotethe mean QTc effect in the control group (Dose 0) and μ1, μ2, μ3 denote the mean QTceffects at Doses 1, 2, and 3, respectively. The linear trend based on an ordinal dosing scaleis examined by testing the following linear contrast:

Contrast of four means = −3μ0 − 1μ1 + 1μ2 + 3μ3.

This trend test is performed in a sequential fashion to identify the highest NOEL. If thelinear contrast of the four means is not significant, one concludes that the high dose (Dose3) is the NOEL and no further testing is performed. If the linear contrast is significant, the

Page 117: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

104 Pharmaceutical Statistics Using SAS: A Practical Guide

test procedure continues to assess the significance of a linear trend in the first three meansusing the following contrast:

Contrast of three means = −1μ0 + 0μ1 + 1μ2.

If this linear contrast is not significant, one concludes that the medium dose (Dose 2) is theNOEL and testing stops. However, if the test is significant, the test procedure continues tocompare the control and low dose (Dose 1):

Contrast of two means = −1μ0 + 1μ1.

If this linear contrast is not significant, the low dose (Dose 1) is the NOEL. Otherwise, theNOEL is not established for this response variable.

5.4.2 Power EvaluationWe evaluate the statistical power of the sequential testing method via simulation. Thefollowing parameters are needed: the number of simulations, the sample size in eachtreatment group, the effect size in each treatment group, and the common variance.

The simulation study will use 2000 simulations with n = 3 per sex or n = 4 per sex ineach treatment group. Let yijk denote the observation for the k-th animal in the i-thtreatment group (i = 0, 1, 2, 3) and j-th sex group (j = 1 denotes males and j = 2 denotesfemales). Suppose that yijk is normally distributed with mean μij and variance σ2.

The dose-response profile is defined to be flat from Dose 0 up to Dose 2 and assumed toshow an increase at Dose 3. In other words,

μ0j = μ1j = μ2j

for j = 1, 2. To allow for a sex difference at Dose 3, we make the following assumptions.The treatment effects in male and female animals at Dose 3 are given by

μ31 = μ01(1 + δ1), μ32 = μ02(1 + δ2).

Here δ1 is the relative treatment difference in the male animals at Dose 3 compared to Dose0 and, similarly, δ2 is the relative treatment difference in the female animals at Dose 3compared to Dose 0. It is worth noting that the assumptions given above result inconservative power estimates. If drug effects were present at Doses 1 and 2, the power ofthe sequential testing method would be greater.

The control means in males and females as well as the variance of QTc interval wereestimated from a historical database including pre-treatment QTc values from 91 male and91 female beagle dogs:

μ01 = 236.35, μ02 = 237.88, σ2 = 102.98.

Power simulations are performed under 12 scenarios defined by 6 combinations of δ1 andδ2 (δ1 = 0.05, δ2 = 0; δ1 = 0.05, δ2 = 0.05; δ1 = 0.1, δ2 = 0; δ1 = 0.1, δ2 = 0.1; δ1 = 0.15,δ2 = 0; δ1 = 0.15, δ2 = 0.15) and two values of the sample size per sex per treatment group(n = 3 and n = 4). Simulated data are generated by calling the %SIMULQT macro thatcan be found on the book’s companion Web site. For example, the following call simulatesQTc interval data for δ1 = δ2 = 0.05 and three animals per sex per treatment group (theparameters of the %SIMULQT macro are defined in the code available on the book’scompanion Web site):

%simulqt(n_sim=2000, avgmale=236.35, avgfemale=237.88, var=102.98,n=3, delta1=0.05, delta2=0.05, out=simul1, seed=2631);

Page 118: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 105

Program 5.3 analyzes the simulated data using the MIXED procedure, with theESTIMATE statements specifying the linear contrasts of treatment means. The p-valuesfor Tests A, B, and C are saved in the TESTS data set. To evaluate the power of Test D,the FINALCOUNT data set converts the three p-values into binary variables (TESTA,TESTB, and TESTC) based on the 0.05 significance level. The TESTD variable is thendefined as the sum of TESTA*TESTC and (1-TESTA)*TESTB. Finally, the binaryvariables are summarized using the MEANS procedure to compute the power of each test.

Program 5.3 Power evaluation in the two-factor model for QT interval

proc mixed data=simul1;by simul;class simul dose sex animal;model qtc=sex|dose;/* Test A */estimate ’Linear trend*sex’ dose*sex 3 -3 1 -1 -1 1 -3 3;/* Test B */estimate ’Combined trend test’ dose -3 -1 1 3;/* Test C */estimate ’Trend test in females’ dose -3 -1 1 3

dose*sex -3 0 -1 0 1 0 3 0;estimate ’Trend test in males’ dose -3 -1 1 3

dose*sex 0 -3 0 -1 0 1 0 3;ods output estimates=tests;

data tests (keep=simul label probt);set tests;if probt<0.0001 then probt=0.0001;

proc transpose data=tests out=testfinal;by simul;var label probt;

data testfinal (drop=_name_ _label_);set testfinal;rename col1=flagp col2=combp col3=femalep col4=malep;if _name_=’Probt’;

data finalcount;set testfinal;retain testA testB testC testD 0;testA=(flagp<0.05);testB=(combp<0.05);testC=max((femalep<0.05),(malep<0.05));testD=testA*testC+(1-testA)*testB;

proc means data=finalcount noprint;var testA testB testC testD;output out=power;

data summary (keep=testA testB testC testD);set power;if _stat_=’MEAN’;

proc print data=summary noobs;format testA testB testC testD 5.3;run;

Output from Program 5.3

testA testB testC testD

0.051 0.421 0.414 0.445

Page 119: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

106 Pharmaceutical Statistics Using SAS: A Practical Guide

Output 5.3 displays the estimated power of the four tests. The power of the overallanalysis (Test D) in this scenario (δ1 = δ2 = 0.05 and n = 3) is clearly too low (44.5%). Theestimated powers for all 12 scenarios are summarized in Table 5.1. The power of thesequential testing procedure is above 95% when Dose 3 is expected to prolong the QTcinterval by 10% in both male and female beagle dogs.

Table 5.1 Estimated Power of Sequential Testing Procedure in the Two-Factor Model for QT Interval

Sample size per sexper treatment groupTreatment effect

in males (δ1)Treatment effectin females (δ2) n = 3 n = 4

0.05 0 23.2% 30.2%0.05 0.05 44.5% 59.2%0.1 0 63.4% 78.7%0.1 0.1 95.7% 98.8%0.15 0 93.2% 98.4%0.15 0.15 100.0% 100.0%

Evaluation of Type I Error Rate in a Two-Factor ANOVA ModelAnother important characteristic of the sequential testing method is the probability of aType I error. The Type I error rates for Tests A and B are 5% and, under an additionalassumption of independent multiple tests, the Type I error rate for Test C is1 − (1 − 0.05)2 = 9.75%. Although calculation of the Type I error rate for Test D is not asstraightforward, it can be evaluated by simulation.

To estimate the Type I error rate associated with Test D, one needs to create asimulated data set under the global null hypothesis of no drug effect (δ1 = δ2 = 0) as shownbelow:

%simulqt(n_sim=2000, avgmale=236.35, avgfemale=237.88, var=102.98,n=3, delta1=0, delta2=0, out=simul1, seed=4641);

The Type I error rate of Test D is computed using Program 5.3.

Output from Program 5.3 (Computation of the Type I error rate)

testA testB testC testD

0.054 0.054 0.098 0.084

Output 5.3 (Computation of the Type I error rate) shows the estimated Type I errorprobabilities of Tests A, B, C, and D. The estimated Type I error rate of the overallanalysis (Test D) is 8.4%. Since this rate is greater than 5%, one can consider adjusting thesignificance levels for Tests A and B downward, i.e., carrying them out at a level that islower than 0.05.

5.5 Statistical Analysis of a One-Factor Design with Repeated MeasuresIn this section, we discuss the methods for analyzing the body weight data from rodent andlarge animal toxicology studies. Since body weights of each animal are collected throughoutthe study, they are repeated measures data. The repeated measures analysis is a logicalchoice for large animal toxicology studies (Thakur, 2000) and rodent toxicology studies

Page 120: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 107

(Hoffman et al., 2002) with data that are repeated in nature. For a typicalfour-treatment-group two-year carcinogenicity study, there could be as many as 60 rats persex for each treatment group, each receiving a 0 (vehicle), low, mid, or high dose of acompound daily for the entire two years. The body weights of each animal are usuallycollected weekly up to 13 weeks and biweekly thereafter. The purpose of the statisticalanalysis is to evaluate compound-related effects on body weight. The statistical methodsare described and the analysis of the data is carried out using SAS. For a detaileddescription of repeated measures models and issues arising in the analysis of incompletedata, we refer the reader to Chapter 12, “Analysis of Incomplete Data.”

5.5.1 Objectives of Statistical ComparisonsThe main interest of the analysis is to identify the NOEL on body weight in each sex bycomparing treatment groups to the vehicle control. Treatment-related effects need to beinterpreted with reference to the time intervals in which they were observed. Thecompound-related effects at a single time point are discussed first to help lay thefoundation for evaluation of those effects within a time span.

5.5.2 Assessment of Compound-Related Effects at a Single Time PointCompound-related effects can be assessed using the sequential trend test described inSection 5.4.1. The trend test is performed to evaluate the monotonic dose-responserelationship in group means. However, if researchers are also interested in detectingstatistically significant differences between the lower doses and the control in the absence ofa high dose effect, then the Dunnett test (Dunnett, 1964) at the significance level of 0.05can be applied as a secondary test. To avoid an inflated Type I error rate by routinelyperforming Dunnett’s t-test in the absence of a high dose effect, researchers can firstperform an F -test at a lower significance level, 0.01. Dunnett’s t-test is performed only ifthe F -test is significant. If the effects detected at lower doses in the absence of a high doseeffect would be dismissed as not being dose-responsive, then neither the F -test norDunnett’s t-test would be considered.

5.5.3 Assessment of Compound-Related Effects within a Time SpanThe evaluation of compound effects can be expanded from a single time point to the entiretime span of the experiment (Hoffman et al., 2002). As the duration of a study increases, sodoes the number of body weight measurements. For example, in a two-year carcinogenicitystudy there would be more than 50 body weight measurements for each animal surviving tothe end of the study. As one expects the growth pattern to change as animals mature, bodyweights are averaged accordingly to capture the characteristics. The calculation of averagebody weights in selected analysis intervals and the handling of missing data due to deathare discussed in this section. The inclusion of baseline and the selection of a covariancestructure for each animal across time are detailed first. After that, statistical methods forevaluation of monotonic and nonmonotonic dose-response relationships are explained.

Calculation of Interval-Averaged Body Weights

Analysis intervals for averaging body weights that are used in rodent and large animalstudies at Eli Lilly and Company are listed in Table 5.2. Statistical analysis of rodent bodyweights collected in the first 3 months of the rapid growth phase is performed in oneanalysis. After 3 months, the growth of a rodent slows down and enters into themaintenance phase. Body weights beyond 3 months are evaluated in the second statisticalanalysis. The baseline body weight of each animal is included in both analyses as acovariate. Large animal studies are typically 2 weeks, or 1, 3, 6, 9, or 12 months induration. Evaluation of large animal body weights collected in the first 6 months is

Page 121: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

108 Pharmaceutical Statistics Using SAS: A Practical Guide

Table 5.2 Analysis Intervals for Rodent and Large Animal Studies

Timea Rodent studies Large animal studies

Weeks 1–5 Every week Every weekWeeks 6–14 Every 2 weeks Every 4 weeksWeeks 15–26 Every 4 weeks Every 4 weeksWeeks 27–104b Every 14 weeks Every 4 weeksaApproximate time intervals which may vary from study to study.bDuration of dog studies is generally up to 52 weeks.

performed in one statistical analysis, while additional body weights collected beyond 6months are analyzed in a second analysis.

Interval-averaged body weights for rodents are derived using the %ONEWEEK and%WEEKINT macros provided on the book’s companion Web site. The INDDATA data setincludes body weight measurements, collection days and general study information. Thefirst few weeks of the study are called the initial period when no averaging is required.Weekly body weights are saved as INT BW for those weeks starting from the study daythat the first weekly data are recorded, WKLY FIRST, to the study day that the lastweekly data are recorded, WKLY END. This is done using the %ONEWEEK macro. Afterthe initial period, body weights in the selected intervals are averaged and saved as INT BWin the macro. To obtain a representative body weight for the selected interval, the initialbody weight in the current interval, which is the last body weight of the previous interval, isalso included in the calculation. The %WEEKINT macro computes INT BW by specifyingthe number of time points for averaging (NUM TIMEPTS), the start day (INT START),and the end day (INT END) of the interval. This macro pulls the last observation from theprevious interval into the calculation of INT BW for the current interval.

As an illustration, Program 5.4 applies the macros to derive five weekly body weightsand four biweekly interval-averaged body weights post dosing as INT BW. These areappended to the base data set, ALL INTERVAL. ALL INTERVAL contains the weeklybody weights for Days -1, 6, 13, 20, 27, and 34, and biweekly interval-averaged bodyweights for Days 48, 62, 76, and 90.

Program 5.4 Calculation of interval-averaged body weights

%oneweek(wkly_first=-1,wkly_end=34);%weekint(num_timepts=2,int_start=41,int_end=90);

Partial output of ALL INTERVAL from Program 5.4

Gender Phase Day Animal Group BW INTVL_BW

Male Treatment -1 1001 1 132.5 132.5Male Treatment 6 1001 1 166.5 166.5Male Treatment 13 1001 1 194.8 194.8Male Treatment 20 1001 1 212.1 212.1Male Treatment 27 1001 1 246.3 246.3Male Treatment 34 1001 1 255.5 255.5Male Treatment 41 1001 1 277.3 NCMale Treatment 48 1001 1 294.7 275.8Male Treatment 55 1001 1 308.8 NCMale Treatment 62 1001 1 317.3 306.9Male Treatment 69 1001 1 330.8 NCMale Treatment 76 1001 1 328.3 325.5Male Treatment 83 1001 1 339.0 NCMale Treatment 90 1001 1 339.8 335.7

Page 122: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 109

Female Treatment -1 2051 2 101.8 101.8Female Treatment 6 2051 2 116.5 116.5Female Treatment 13 2051 2 127.5 127.5Female Treatment 20 2051 2 145.5 145.5Female Treatment 27 2051 2 152.1 152.1Female Treatment 34 2051 2 156.0 156Female Treatment 41 2051 2 162.6 NCFemale Treatment 48 2051 2 171.9 163.5Female Treatment 55 2051 2 178.6 NCFemale Treatment 62 2051 2 189.5 180Female Treatment 69 2051 2 196.5 NCFemale Treatment 76 2051 2 197.8 194.6Female Treatment 83 2051 2 201.9 NCFemale Treatment 90 2051 2 204.2 201.3

Handling of Missing Data Due to Death

As animals age, their mortality rate also increases. It is not unusual to see 50% of theanimals die before the end of a carcinogenicity study. Missing data due to death aredifferent from missing data due to other reasons. When averaging body weights for ananimal in a selected time interval, a missing value will be assigned in the former case whilean average would be calculated based on all available body weights in the latter case. Allbody weights up to the time interval of death are included in the statistical analysis. Bodyweights that are collected prior to death but that are not included in the statisticalanalysis would be incorporated into the overall assessment of mortality data.

Program 5.5 demonstrates how to properly handle the missing data. For each animal, asurvival index (SURVIVAL DAY) is created to indicate the last day of non-missingobservations. This index is used to identify if a missing body weight is due to death orother reasons. If this survival index day falls within an interval, the derived interval bodyweight (INT BW) is set to missing for that animal. Otherwise, the program averages allavailable non-missing body weights.

The following arguments are used in Program 5.5:

• SURVIVAL DAY is the last day of non-missing body weights.• START is the first day of an interval (e.g., Day 48).• END is the last day of an interval (e.g., Day 69).

Program 5.5 Handling of missing data

data survival_index;set all;if bw ne .;

proc sort data=survival_index;by animal gender;

data survival_index;set survival_index;by animal gender day;if last.animal;survival_day=day;keep animal gender survival_day;

data week&end;merge week&end(in=in1) survival_index(in=in2);by animal;if in1 and in2;if &start<=survival_day<&end then int_bw=.;run;

Page 123: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

110 Pharmaceutical Statistics Using SAS: A Practical Guide

The output of Program 5.5 can be summarized into four possible scenarios. Toillustrate, Table 5.3 includes both the weekly body weights and the derived interval bodyweights. For the first month, INT BW values were captured directly from the five weeklybody weights. INT BW ending on Day 48 was computed by averaging the body weightscollected on Days 34, 41, and 48. For the last time interval that ended on Day 62, INT BWis derived from the average of three time points: Days 48, 55, and 62. The followingscenarios are presented in Table 5.3:

Scenario 1. Animal 1001 did not have any missing values.Scenario 2. Animal 1002 had a missing value on Day 55. Hence INT BW for Day 62 was

computed by averaging the body weights from Days 48 and 62.Scenario 3. Animal 1003 had two missing values on Days 41 and 48. Hence INT BW for

Day 48 was computed from Day 34, and INT BW for Day 62 was computedby averaging the body weights from Days 55 and 62.

Scenario 4. Animal 1004 is assumed dead right after Day 48 since there were no datacollected beyond Day 48. Therefore, INT BW for the last interval was set tobe missing.

Table 5.3 Examples of INT BW Computation and Missing Data Handling

Study dayAnimalID Parameter 6 13 20 27 34 41 48 55 62

1001 BW 166.5 194.8 212.2 246.3 277.3 294.7 308.8 317.3 330.8INT BW 166.5 194.8 212.2 246.3 277.3 293.6 319.0

1002 BW 166.5 194.8 212.2 246.3 277.3 294.7 308.8 . 330.8INT BW 166.5 194.8 212.2 246.3 277.3 293.6 319.8

1003 BW 166.5 194.8 212.2 246.3 277.3 . . 317.3 330.8INT BW 166.5 194.8 212.2 246.3 277.3 277.3 324.1

1004 BW 166.5 194.8 212.2 246.3 277.3 294.7 308.8 . .INT BW 166.5 194.8 212.2 246.3 277.3 293.6 .

5.5.4 Selection of the Covariance Structure for Each AnimalTo account for the initial differences in baseline body weight, it is important to include itas a covariate in the statistical model. In addition, to account for the correlation amongrepeated measures from the same animal, one needs to select an appropriate covariancestructure for each animal. As a default covariance structure, consider repeated measuresdata from a split-plot design with a treatments, b time points, and c animals. The split-plotmodel (Aldworth and Hoffman, 2002) with treatment as the whole-plot factor, time as thesubplot factor, and baseline as a covariate is

yijk = μ + αi + γ(xik − x..) + dk(i) + βj + (αβ)ij + eijk,

where i = 1, . . . , a, j = 1, . . . , b and k = 1, . . . , c. In this equation, y is the body weight, μ isthe grand mean, α is the treatment effect, γ is the regression coefficient for the covariate xand the overall mean of the covariate x, β is the time effect, αβ is the treatment by timeinteraction, d is the random error for the animal nested in the treatment, and e is randomerror at each time point.

The covariance matrix for an animal in this model has the variance componentsstructure (VC). The covariance terms in VC are nonnegative. If negative correlations are

Page 124: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 111

allowed in the covariance structure of VC, the matrix will have a compound symmetricstructure (CS).

In addition, the variances of an animal at different time points could be heterogeneous.Therefore, one can consider other covariance structures, including homogeneous variancecomponents (VC), heterogeneous variance components (UN(1)), homogeneous compoundsymmetry (CS), heterogeneous compound symmetry (CSH) and spatial power (SP[POW]).

For the UN(1) and SP(POW) structures, both REPEATED and RANDOM statementsare included in the MIXED procedure. An animal is specified as the subject in bothstatements and the intercept is specified in the RANDOM statement as a random effect.

For the CS and CSH structures, only the REPEATED statement is included while forthe VC structure, only the RANDOM statement is included. The finite-sample correctedAkaike’s Information Criterion (Keselman et al., 1998) can be used for selecting thecovariance structure. The Kenward and Roger method (Kenward and Roger, 1997) isgenerally recommended for the denominator degrees of freedom. The PROC MIXEDsyntax for each covariance structure discussed above follows:

Homogeneous variance components (VC)

proc mixed data=one;class trt time animal;id trt time animal;model body_wt=trt time trt*time baseline/ddfm=kenwardroger solution;random int/type=vc subject=animal s;

Heterogeneous variance components (UN(1))

proc mixed data=one;class trt time animal;id trt time animal;model body_wt=trt time trt*time baseline/ddfm=kenwardroger solution;repeated time/type=un(1) subject=animal r;random int/subject=animal s;

Note that TYPE=UN(1) can be replaced with TYPE=VC GROUP=TIME.

Homogeneous compound symmetry (CS)

proc mixed data=one;class trt time animal;id trt time animal;model body_wt=trt time trt*time baseline/ddfm=kenwardroger solution;repeated time/type=cs subject=animal r;

Heterogeneous compound symmetry (CSH)

proc mixed data=one;class trt time animal;id trt time animal;model body_wt=trt time trt*time baseline/ddfm=kenwardroger solution;repeated time/type=csh subject=animal r;

Page 125: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

112 Pharmaceutical Statistics Using SAS: A Practical Guide

Spatial power (SP[POW])

proc mixed data=one;class trt time animal;id trt time animal;model body_wt =trt time trt*time baseline/ddfm=kenwardroger solution;repeated time/type=sp(pow) (time) subject=animal r;random int/subject=animal s;

Evaluation of Monotonic and Nonmonotonic Dose-Response Relationships

A repeated measures analysis of variance is performed to assess compound-related effectsacross time. Treatment and time are entered in the statistical model as the two mainfactors. The interaction between treatment and time is also included in the model. Basedon the significance of the interaction, compound effects are assessed either at each timepoint when the interaction is significant, or on results pooled across the entire time spanwhen the interaction is not significant. This approach allows researchers to detect the startand end of compound-related effects.

The assessment of compound-related effects for repeated measures analysis of variance iscarried out in the same manner as for single-time measurements. The additional dimensionof time requires more detailed specifications for the comparisons of treatment groups atselected time points. The basic concept of evaluating a monotonic dose-responserelationship using the sequential trend test by Tukey, Ciminera and Heyse (1985) andsupplementing it with a pairwise to control test for a nonmonotonic dose-responserelationship remains the overall strategy.

To test for a monotonic dose-response relationship in treatment, one first needs toevaluate the interaction between time and treatment by performing the following threeinteraction tests:

• Test 1 (linear trend in treatment by time at the 0.01 significance level). This testevaluates the similarity of the monotonic dose-response relationship across time.Consider a slope for each dose-response relationship being evaluated. The test checks forequality of these slopes at all time points.

• Test 2 (linear trend in treatment by linear trend in time at the 0.05 level). This testchecks if the slopes defined above change consistently across time, e.g., if they continueto rise or continue to drop.

• Test 3 (linear trend in treatment by quadratic trend in time at the 0.05 level). This testchecks if the slopes change in a quadratic fashion, i.e., rising or dropping in onedirection to a point and then changing directions for the rest of the time span.

Tests 2 and 3 help researchers understand the treatment related dose-response effectswithin a time span. The interaction between the linear trend in treatment and time is amore general catch-all test to capture those scenarios when the slopes of the monotonicdose-response vary significantly across time and the profile of the slopes may be morecomplicated than linear or quadratic.

Following the strategy for handling monotonic and nonmonotonic treatment effects at asingle time point, Test 1 is carried out at the 0.01 level and Tests 2 and 3 are carried out atthe 0.05 level. If any of the three interaction tests is statistically significant at therespective significance level, contrasts need to be specified to examine the treatment effectseither on the results pooled across time, or at each time point in addition.

In order to evaluate the monotonic dose-response relationship in treatment and thetreatment by time interaction terms, several ESTIMATE and CONTRAST statementsneed to be specified. The %REPMEAS49 macro provided on the book’s companion Web

Page 126: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 113

site is an example of proper ESTIMATE and CONTRAST parameters in a study design offour groups with nine time points post dosing. The doses are measured on an ordinal scale.

If no monotonic dose-response relationship is detected in treatment and a lower doseeffect in the absence of a high dose effect may be meaningful, one can evaluate anonmonotonic dose-response relationship. In the one-factor design, this is accomplished byconducting an F -test at a lower significance level, 0.01, and carrying out Dunnett’s t-testat the 0.05 level only after the F -test is shown to be significant.

In the repeated measures analysis based on PROC MIXED, Dunnett’s t-test wouldcompare all combinations of treatment and time back to the first time point of the controlgroup. This is not the same as comparing the treated group means back to the control ateach time point. Therefore, Dunnett’s t-test is replaced with the Bonferroni-adjusted t-testat the 0.05 level. The Bonferroni-adjusted t-test is preceded by two F -tests carried out atthe 0.01 level for (1) the treatment by time interaction and (2) the treatment main effect:

• If the treatment by time interaction is significant, the Bonferroni-adjusted t-test will beapplied to the treatment means for each time point.

• If the treatment main effect alone is significant, the Bonferroni-adjusted t-test will beapplied only to the treatment means pooled across all time points.

• If neither is significant, no further testing is performed.

Note that the Bonferroni-adjusted t-test can be carried out using the %BONF ADJmacro given on the book’s companion Web site.

SAS Module for the Evaluation of Monotonic and Nonmonotonic Dose-ResponseRelationships

A comprehensive and flexible SAS module was developed by the authors to carry out thestatistical analysis for the evaluation of monotonic and nonmonotonic dose-responserelationships in a repeated measures ANOVA framework. In this macro-parameter-drivenmodule, users can specify the names of the analysis variables and covariates (if any), levelsof time factor, the covariance structure, denominator degrees of freedom method (DDFM),and an option of yes/no for inference tests. As an illustration, SAS code provided on thebook’s companion Web site uses the macro %PRF1FRM to analyze the INDDATA dataset. Results from the analysis of the INDDATA data set using the module are given inTables 5.4 and 5.5.

5.6 SummaryThe growing complexity of guideline studies, and the increasing number of measurementsrequired therein, present major challenges to nonclinical scientists and statisticians. Centralto defining hazards and subsequently assessing risk is a clear understanding of the studydesign and data characteristics. This chapter briefly defines statistical aspects of toxicologystudies with examples implemented using SAS. We have described randomization schemesfor two commonly used designs in toxicology, namely, the parallel design and Latin squaredesign. We have presented the statistical power evaluation of the heart-rate corrected QTintervals from a large animal toxicology study. We have also discussed the statisticalanalysis of treatment-related effects on body weights from general toxicology studies. Fromcreating randomization schemes, calculating the power of a test, performing statisticalanalysis and finally to reporting results in nicely formatted tables, SAS has been anindispensable tool for nonclinical statisticians in the pharmaceutical industry.

Page 127: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

114 Pharmaceutical Statistics Using SAS: A Practical Guide

Table 5.4 Summary Table for Body Weight Data Using a Repeated Measures Analysis of Variance, Days 6–48

Repeated Measures ANOVA : Compound - TEST STUDY Study - int parms BW testAnalysis Variable : Interval BW (g)

Baseline (Day -1) was used as covariateGENDER = Female

DayGroup Statistics Baseline Overall Day 6 Day 13 Day 20 Day 27 Day 34 34–48

1 Mean 100.39 159.22 119.51 130.98 145.51 153.62 160.55 168.32SD 4.34 NA 5.23 5.22 7.55 8.11 7.41 8.09N 15 15 15 15 15 15 15 15LSM NA 159.27 119.56 131.03 145.56 153.67 160.60 168.36LSM s.e. NA 1.42 1.66 1.66 1.66 1.66 1.66 1.66

2 Mean 100.25 162.23 118.12 131.64 147.95 156.39 162.15 170.54SD 4.36 NA 5.32 6.00 5.94 7.42 7.40 7.67N 15 15 15 15 15 15 15 15Mean: % Chg from Cntrl 0 2 -1 1 2 2 1 1LSM NA 162.41 118.30 131.82 148.13 156.57 162.33 170.72LSM s.e. NA 1.42 1.66 1.66 1.66 1.66 1.66 1.66Trend p-val# NT 0.124 NT NT NT NT 0.462 0.317

3 Mean 100.49 152.19 117.95 129.25 142.09 151.23 155.19 160.28SD 4.60 NA 5.27 5.03 5.79 5.92 7.03 7.21N 15 15 15 15 15 15 15 15Mean: % Chg from Cntrl 0 -4 -1 -1 -2 -2 -3 -5LSM NA 152.14 117.90 129.20 142.03 151.17 155.13 160.22LSM s.e. NA 1.42 1.66 1.66 1.66 1.66 1.66 1.66Trend p-val# NT 0.001* NT NT 0.135 NT 0.022* 0.001*

4 Mean 100.61 154.34 117.62 129.28 142.68 151.04 155.15 161.36SD 4.27 NA 5.88 6.01 7.24 7.75 7.84 7.20N 15 15 15 15 15 15 15 15Mean: % Chg from Cntrl 0 -3 -2 -1 -2 -2 -3 -4LSM NA 154.17 117.45 129.11 142.51 150.87 154.97 161.18LSM s.e. NA 1.42 1.66 1.66 1.66 1.66 1.66 1.66Trend p-val# NT <.001* 0.364 0.260 0.042* 0.065 0.002* <.001*

ALL Trt F-test p-val++ <.001*

INTN Trt*Time p-val++ <.001*LinTrt*Time p-val++ <.001*LinTrt*LinTime p-val+ <.001*LinTrt*QdrTime p-val+ 0.698

# : Level of significance tested = .05; Two-sided test. NT : Not tested.++ : Level of significance tested = .01. NA : Not applicable.+ : Level of significance tested = .05. KENWARDROGER was used for the DDFM.* : Statistically significant. CS covariance structure over time was selected for the model.

Page 128: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 5 Some Statistical Considerations in Nonclinical Safety Assessment 115

Table 5.5 Summary Table for Body Weight Data Using a Repeated Measures Analysis of Variance, Days48–90

Repeated Measures ANOVA : Compound - TEST STUDY Study - int parms BW testAnalysis Variable : Interval BW (g)

Baseline (Day -1) was used as covariateGENDER = Female

Day Day DayGroup Statistics 48–62 62–76 76–90

1 Mean 178.38 185.52 190.61SD 9.99 11.50 12.82N 15 15 15LSM 178.43 185.56 190.67LSM s.e. 1.66 1.66 1.66

2 Mean 182.91 192.63 197.70SD 8.72 9.37 9.26N 15 15 15Mean: % Chg from Cntrl 3 4 4LSM 183.09 192.82 197.88LSM s.e. 1.66 1.66 1.66Trend p-val# 0.049* 0.003* 0.003*

3 Mean 166.55 171.59 175.60SD 8.08 8.86 8.87N 15 15 15Mean: % Chg from Cntrl -7 -8 -8LSM 166.49 171.54 175.54LSM s.e. 1.66 1.66 1.66Trend p-val# <.001* <.001* <.001*

4 Mean 170.20 178.28 183.45SD 7.21 7.98 8.06N 15 15 15Mean: % Chg from Cntrl -5 -4 -4LSM 170.03 178.11 183.27LSM s.e. 1.66 1.66 1.66Trend p-val# <.001* <.001* <.001*

ALL Trt F-test p-val++

INTN Trt*Time p-val++LinTrt*Time p-val++LinTrt*LinTime p-val+LinTrt*QdrTime p-val+

# : Level of significance tested = .05; Two-sided test. NT : Not tested.++ : Level of significance tested = .01. NA : Not applicable.+ : Level of significance tested = .05. KENWARDROGER was used for the DDFM.* : Statistically significant. CS covariance structure over time was selected for the model.

AcknowledgmentsThe authors wish to thank Dr. Gheorghe Doros and Mr. James Hoffman for reviewing andrefining the chapter.

ReferencesAldworth, J., Hoffman, W.P. (2002). “Split-plot model with covariate: A cautionary tale.” The

American Statistician. 56, 284–289.Box, G.E.P., Hunter, W.G., Hunter, J.S. (1978). “Designs with more than one blocking variable.”

Statistics for experimenters. New York: Wiley. 245–280.Chiang, A.Y., Smith, W.C., Main, B.W., Sarazan, R.D. (2004). “Statistical power analysis for

hemodynamic cardiovascular safety pharmacology studies in beagle dogs.” Journal ofPharmacological and Toxicological Methods. 50, 121–130.

Dunnett, C.W. (1964). “New tables for multiple comparisons with a control.” Biometrics. 20,482–491.

Fridericia, L.S. (1920). “Die Systolendauer im Elecktrokardiogramm bei normalen Menschen undbei Herzkranken.” Acta Medica Scandinavia. 53, 469–486.

Page 129: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

116 Pharmaceutical Statistics Using SAS: A Practical Guide

Hoffman, W.P., Ness, D.K., van Lier, R.B. (2002). “Analysis of rodent growth data in toxicologystudies.” Toxicological Sciences. 66, 313–319.

International Conference on Harmonization of Pharmaceuticals for Human Use (ICH). (2004). S7B:“The nonclinical evaluation of the potential for delayed Ventricular repolarization (QT intervalprolongation) by Human Pharmaceuticals. Step 2 Revision.” Available at http://www.ich.org.

Kenward, M.G., Roger, J.H. (1997). “Small sample inference for fixed effects from restrictedmaximum likelihood.” Biometrics. 53, 983–997.

Keselman, H.J., Algina, J., Kowalchuk, R.K., Wolfinger, R.D. (1998). “A comparison of twoapproaches for selecting covariance structures in the analysis of repeated measurements.”Communications in Statistics. Simulation and Computation. 27, 591–604.

Kinter, L.B., Siegl, P.K.S., Bass, A.S. (2004). “New preclinical guidelines on drug effects onventricular repolarization: Safety pharmacology comes of age.” Journal of Pharmacological andToxicological Methods. 49, 153–158.

Lin, K.K. (2001). Guidance for industry statistical aspects of the design, analysis, andinterpretation of chronic rodent carcinogenicity studies of pharmaceuticals (draft guidanceonline). Rockville: US Department of Health and Human Services, Food and DrugAdministration, Center for Drug Evaluation and Research (CDER). Available athttp://www.fda.gov/cder/guidance/815dft.pdf.

Thakur, A.K. (2000). “Statistical issues in large animal toxicology.” International Journal ofToxicology. 19, 133–140.

Tukey, J.W., Ciminera, J.L., Heyse, J.F. (1985). “Testing the statistical certainty of a response toincreasing doses of a drug.” Biometrics. 41, 295–301.

Page 130: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6

Nonparametric Methods inPharmaceutical Statistics

Paul Juneau

6.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117

6.2 Two Independent Samples Setting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 118

6.3 The One-Way Layout . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .129

6.4 Power Determination in a Purely Nonparametric Sense . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149

Nonparametric, or distribution-free, statistical methods are very useful in the setting ofpharmaceutical research. These methods afford data analysts the ability to relax some ofthe assumptions typically made by their Gaussian (normality-based) analogues. In somesettings (e.g., drug discovery investigations), these assumptions may not be verifiable dueto small sample sizes. In others, where larger sample sizes are employed (e.g., clinical trialsettings), the assumption of a Gaussian (normal) distribution is not met because of thepresence of heavy-tails in measurement response or a large degree of skewness. This chaptercovers two settings found commonly in pharmaceutical research (two-sample setting andone-way layout) and discusses sample size determination in a nonparametric sense. Theintroduced statistical methods are illustrated using examples from drug discovery studiesand clinical trials.

6.1 IntroductionWhy are nonparametric statistical methods important to pharmaceutical research? Toreally address this question, we first must begin by looking at the nature of pharmaceuticalresearch. The goal of pharmaceutical research is to discover and develop new medicinesthat are able to cure or moderate various disease symptoms or conditions. The diseaseresponse (i.e., physiological trauma) can vary greatly from individual to individual becauseof a complex inherent genetic variability. The corresponding statistical phenomenonassociated with this genetic variation to disease response is that measurements collected onsuch a cohort can be skewed by the extreme response of a small portion of the individualsunder consideration.

Consider, as an example, the search for a new anti-inflammatory agent. Inflammatoryresponse (e.g., swelling) can vary greatly in untreated subjects. Some individuals can havea very extreme response to a biological insult that produces an inflammatory response,

Paul Juneau is Associate Director, Nonclinical Statistics, Pfizer, USA.

Page 131: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

118 Pharmaceutical Statistics Using SAS: A Practical Guide

causing the distribution of those responses to be very “heavy” or “long-tailed” in the upperdirection (e.g., a great deal of swelling). The common summary statistic for continuousresponses in such a setting, the mean, is greatly influenced by the presence of a smallnumber of high responses and “over-represents” the location of the untreated subjects’distribution (we will see that a similar phenomenon can also occur in the study of novelantibacterial drugs).

A similar phenomenon can occur, but this time in treated subjects, in the study of thetoxicology of new agents. The genetic variability of individuals can vary greatly as thebody responds to higher and higher levels of a compound. Some subjects in a high dosegroup of a toxicology study may exhibit very high or very low responses that skew thedistribution. In both cases (the study of efficacy and toxicology), it becomes apparent thattraditional Gaussian (normal) statistical methods may fail because of the requiredassumption of symmetry. Nonparametric, or distribution-free, statistical methods liberateus from the need to assume symmetry in response. We no longer impose an assumption onour inference that may not be verifiable (because of small sample sizes) or observable.

This chapter is an introduction to popular nonparametric statistical methods in theanalysis of studies in the pharmaceutical industry. It provides a review of some newstatistical methods for inference and an introduction to some fairly infrequently usedtechniques that have existed “below the radar” for several years.

Section 6.2 will be devoted to the two-sample setting, exploring both the equal andunequal dispersion cases. Section 6.3 will discuss aspects of the one-way layout andassociated multiple comparison procedures. Section 6.4 will introduce one approach tosample size determination in a “nonparametric” sense, using data from pilot studies. Eachsection also contains a mixture of SAS/STAT procedures and some original macros toperform elementary nonparametric data analyses.

Throughout this chapter, the discussion of topics will be motivated by providing somereal or simulated examples of data from studies in medical research. In either situation, thepurpose of the example is not to make a medical claim (efficacy or safety of an anonymouscompound) but to illustrate a feature of data critical to the outcome of the statisticalinference.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

6.2 Two Independent Samples SettingAn experiment involving two independent groups is one of the most elementary types ofinvestigations in scientific research, yet is no less valuable than many other morecomplicated multifactor or multilevel experiments. Typically, subjects are randomlyassigned to one of two groups, a treatment is applied, and a measurement is collected on allsubjects in both groups. It is then usually the desire of the investigators to ascertainwhether sufficient evidence exists to declare the two groups to be different. Although manyare familiar enough with statistics to perform two sample comparisons, often, keyassumptions are swept under the rug, as it were, and as a result, errors in inferencesometimes occur.

This section will begin with a review of the assumptions necessary to perform atwo-sample comparison and then discuss a common means to compare the locationparameters of two groups (Section 6.2.2). Section 6.2.3 will discuss one possible solution forthe problem of unequal spread (heteroscedasticity) between two groups. One importantfalse notion about the rank transform and its effect on groups with unequal dispersion willalso be discussed. This section will include examples of data from two clinical trials.

Page 132: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 119

6.2.1 A Review of the Two-Sample SettingWe will begin this section with a slight bit of mathematical formality. Suppose that we havetwo random samples, denoted X1, . . . , Xn1 and Y1, . . . , Yn2 . We assume that the X’s and Y ’scome from a distribution, say, F . Typically, the two-sample problem is characterized by adesire to perform an inference about a shift in location between the distributions of the twosamples. Suppose FX = F (μ) is the distribution of the X’s (with location parameter μ) andFY = F (μ + δ) is the distribution of the Y ’s, shifted by δ. What can be said about ameasure δ, where FX(μ) = FY (μ + δ)? Is δ “reasonably close” to zero or not? If the X’sfollow a Gaussian (normal) distribution, say, N(μ1, σ

2), and the Y ’s are distributed fromanother Gaussian distribution, say, N(μ2, σ

2), we could think of the problem as consideringwhether μ1 = μ2 + δ. In this classic “normal” case, it is very important to note that weassume that the variance of the two Gaussian distributions is “reasonably” the same (oftenreferred to as a common σ2). If latter is not true, we are working under the conditions of afamous setting in statistics, the Behrens-Fisher problem, and we need to make somechanges to our inferential procedures. In either case, some form of a Student’s t-statisticmay be used to compare the locations (means) of the two distributions.

Do the measurements we collect come from a Gaussian distribution? In drug discoverysettings in the pharmaceutical industry, sample sizes in some investigations can be verysmall (total sample size for a study less than 12) and, thus, it might be very difficult toestablish the form of a measurement’s distribution. Moreover, as little is known in thediscovery stages of research about the impact of a compound on a complicated mammalianphysiology, a seemingly outlying or extreme value may in fact be part of the tail of a highlyskewed distribution. With small sample sizes and limited background information, it reallyis anyone’s best guess as to the parametric form of a measurement’s distribution.

What if the distributions are not Gaussian? The standard Student’s t-test will performat the designated level of statistical significance. In most cases, departures from a Gaussiandistribution do not greatly affect the operating characteristics of inferential procedures ifthe two distributions have mild skewness or kurtosis. However, moderate to high levels ofskewness can influence the power of a comparison of two location parameters (Bickel andDocksum, 1977) and, thus, influence our ability to identify potentially efficacious or toxiccompounds.

Shortly, we will look at an example of a two-sample setting where the data aresufficiently skewed to warrant the introduction of a distribution-free test that serves as acompetitor to the standard Student’s two-sample t-test. First, however, it is necessary todescribe a setting where such data could be found: asthma treatment research.

EXAMPLE: Asthma Clinical TrialAsthma is a disease characterized by an obstruction of airflow in the lungs, and thusreferred to as an obstructive ventilatory defect (National Asthma Council Australia, 2002).One of the chief measurements used to characterize the degree of disease is the ForcedExpiratory Volume in 1 second, or FEV1. A patient will exhale into a device (a spirometer)that will measure his or her volume of expired air.

Physicians working on developing new drugs for treatment of asthma are interested in ameasure that characterizes the degree of reversibility of the airflow obstruction. Thus, apatient’s baseline FEV1 will be measured before treatment (at baseline). Typically, 10-15minutes after the administration of a treatment (e.g., a beta2 agonist bronchodilator),FEV1 will be measured a second time. The percent improvement in FEV1 from baseline isused to measure the effectiveness of a new drug:

Percent improvement =Post-treatment FEV1 − Baseline FEV1

Baseline FEV1100%.

Page 133: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

120 Pharmaceutical Statistics Using SAS: A Practical Guide

A medically important improvement is typically at least 12%; however, not allresearchers in the field accept this level.

Consider a simple trial to establish the efficacy of a new treatment for asthma. Sevenhundred patients are randomized to one of two groups. Patients randomized to the firstgroup had baseline FEV1 measurements collected, then received a placebo treatment and,at 10 minutes post-treatment, had a second FEV1 measurement taken. The procedure wassimilar for the second group of patients, except that after measurement at baseline, theyreceived a novel treatment. The percent changes in FEV1 collected in this study areincluded in the SPIRO data set that can be found on the book’s companion Web site.Table 6.1 provides a summary of the results in the two treatment groups and Figure 6.1displays a box plot summary of percent changes in FEV1.

Table 6.1 Summary of Percent Changes in FEV1 in the Asthma Clinical Trial

Group Sample size Mean Standard deviation Median Interquartile range

Placebo 347 9.58 30.89 14.00 37.55Treated 343 13.75 20.08 14.14 29.16

Figure 6.1 Results of the asthma clinical trial

Placebo Treated

-150

-100

-50

0

50

Per

cent

cha

nge

in F

EV

1

Group

The box plots and summary statistics provide some interesting insight into the results ofthe study. First, it appears that for a small number of patients (10 total) a full percentchange score was unavailable. Also, it appears that the placebo group included somepatients whose results were dramatically worse between the initial baseline FEV1 and thelater post-treatment measurement (note the large number of outliers represented by dotsoutside the fence in the box plot). These results produce a long downward tail in thedistribution of change from baseline values and contribute to skewing of the distribution.This also can be observed by the difference of 4 percentage points between the mean andmedian for this group. The level of dispersion, or spread, of the two groups is not identical;however, it is not greatly different. How do we compare the location parameters of twodistributions in the presence of equal, or similar, dispersion?

Page 134: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 121

6.2.2 Comparing the Location of Two Independent Samples I:Similar Dispersion

Returning to the notation introduced previously in Section 6.2.1:

• Group 1: X1, . . . , Xn1 with Xi ∼ F (μ), i = 1, . . . , n1.• Group 2: Y1, . . . , Yn2 with Yj ∼ F (μ + δ), j = 1, . . . , n2.

Here F (λ) is a continuous distribution with location parameter, λ.

Wilcoxon Rank Sum Test

The inference of interest is H0 : δ = 0 versus HA : δ > 0. This null hypothesis can be testedusing the Wilcoxon Rank Sum test (Wilcoxon, 1945). To carry out this test, we begin byperforming a joint ranking of the X’s and Y ’s from the smallest value to the largest. If twoor more values are tied, their ranks are replaced by the average of the tied ranks. TheWilcoxon, W , is simply the sum of the corresponding ranks for the Y ’s, i.e., W =

∑n2j=1 Rj ,

where Rj is the rank of Yj in the combined sample. We reject H0 : δ = 0 at an α level ifW ≥ cα, for a cutoff value, cα, that corresponds to the null distribution of W .

If the minimum of n1 and n2 is sufficiently large, an approximate procedure may beemployed. The Wilcoxon Rank Sum statistic specified above requires some modifications sothat the Central Limit Theorem will hold and the statistic will be asymptotically normal:

WL =(

W − n1(n2 + n1 + 1)2

)/√

Var(WT ).

What is the form of the variance of the W statistics for tied observations, i.e., Var(WT )?Suppose that for all n1 + n2 observations, tied groups of size tv (1, 2, . . . , v, . . . , ξ) exist. Thevariance in the denominator of the statistic is as follows (Hollander and Wolfe, 1999):

Var(WT ) =n1n2

12

(n1 + n2 + 1 −

∑ξv=1(tv − 1)tv(tv + 1)

(n1 + n2)(n1 + n2 − 1)

).

The large sample version of the one-sided hypothesis test is then: Reject H0 : δ = 0 at an αlevel if WL ≥ zα, where zα is the 100(1 − α) quantile from a standard normal distribution.

Before we embark on a discussion of the lower-tailed and two-sided Wilcoxon Rank Sumtests, let’s return to the asthma clinical trial example. Program 6.1 uses the NPAR1WAYprocedure to perform the Wilcoxon Rank Sum test using the SPIRO data set. TheWilcoxon Rank Sum test is requested by the WILCOXON option. PROC NPAR1WAYautomatically uses tied ranks and performs the appropriate adjustment to the test statisticbased upon the large sample form of the statistic. The ODS statement is included to selectthe relevant portion of the procedure’s output. The CLASS statement identifies theclassification or grouping variable. The VAR statement identifies the response variable(percent change in FEV1).

Program 6.1 Wilcoxon Rank Sum test in the asthma trial example

proc npar1way data=spiro wilcoxon;ods select WilcoxonTest;class group;var fevpc;run;

Page 135: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

122 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 6.1

Wilcoxon Two-Sample Test

Statistic 120620.0000

Normal ApproximationZ 0.8071One-Sided Pr > Z 0.2098Two-Sided Pr > |Z| 0.4196

t ApproximationOne-Sided Pr > Z 0.2099Two-Sided Pr > |Z| 0.4199

Z includes a continuity correction of 0.5.

We conclude from Output 6.1 that a statistically significant improvement from baselineFEV1 did not exist at the 0.05 level of statistical significance (one-tailed p-value based onthe normal approximation is 0.2098).

Student’s Two-Sample t-Test

What about other methods of data analysis? Commonly, in the face of skewed data,analysts will use some form of transformation. A commonly used transformation is thenatural logarithm transformation applied to each of the responses. In the exampleillustrated previously, a log or power transformation will not work, as negative values existin the data. Suppose we chose to ignore the long tail of the placebo group’s distribution andperformed a standard Student’s two-sample t-test (Student, 1908). Program 6.2 performsStudent’s two-sample t-test using PROC TTEST. The code to perform a Student’stwo-sample t-test is very similar to the PROC NPAR1WAY code. As in Program 6.1, theODS statement is included to select the relevant portion of the procedure’s output.

Program 6.2 Student’s two-sample t-test in the asthma trial example

proc ttest data=spiro;ods select ttests;class group;var fevpc;run;

Output from Program 6.2

T-Tests

Variable Method Variances DF t Value Pr > |t|

fevpc Pooled Equal 688 -2.10 0.0363fevpc Satterthwaite Unequal 595 -2.10 0.0360

If we select the two-tailed p-value from Output 6.2 (assuming unequal variances in thetwo groups) and adjust it so that it reflects a one-tailed t-test in (p = 0.036/2 = 0.018), wewill infer that the mean FEV1 percent change score was statistically significantly increasedby the new treatment. The long tail of extreme low responses in the control groupinfluenced this conclusion. The extreme negative FEV1 percent change scores did notadversely affect the Wilcoxon Rank Sum test.

Page 136: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 123

Lower- and Two-Tailed Versions of the Wilcoxon Rank Sum TestWe will now move on to complete this section with a brief discussion of the lower-tailedand two-tailed versions of the Wilcoxon Rank Sum test. If the inference of interest isH0 : δ = 0 versus HA : δ < 0, we will reject the null hypothesis at level α if

W ≤ n1(n2 + n1 + 1) − cα,

where cα is the same cutoff mentioned in the first part of this section for the upper-tailedtest. Similarly, for the two-tailed test, if the testing problem is H0 : δ = 0 versus HA : δ �= 0,the null hypothesis is rejected if

W ≤ n1(n2 + n1 + 1) − cα/2 or W ≥ cα/2.

For the large-sample approximate procedures, we will reject the null hypothesis at level α ifWL ≤ zα (upper-tailed test) or |WL| ≥ zα/2 (two-tailed test), where WL is the large-sampleWilcoxon statistic (Hollander and Wolfe, 1999).

6.2.3 Comparing the Location of Two Independent Samples II:Comparisons in the Presence of Unequal Dispersion

EXAMPLE: Antibacterial Clinical TrialLet’s begin our discussion by examining a very simple clinical trial design from clinicalantibacterial research. Bacteria-resistant infections are becoming a serious public healththreat in many parts of the world (World Health Organization, 2002). Consider an agentthat is believed to show some effect against bacteria in an infection that is now thought tobe resistant to most common antibacterial agents. Infected patients are randomized to oneof two groups: a placebo group or a new treatment group. Originally, 400 subjects wererandomized to one of the two arms of the study. Forty-eight hours after receiving treatment,5–10 milliliters of blood were drawn aseptically from each subject to determine his or herblood bacterial count (BBC) in colony-forming units/ml of blood (CFU/ml). Due to someclinical complications during the study, the resultant sample sizes were 184 and 109 (newtherapy and placebo, respectively). The blood bacterial count data collected in the studyare included in the BBC data set that can be found on the book’s companion Web site.

Table 6.2 Summary of Blood Bacterial Count Data in the Antibacterial Clinical Trial

Group Sample size Mean Standard deviation Median Interquartile range

Placebo 184 60.5 92.8 8.0 88.0Treated 109 19.5 38.4 8.0 12.0

Table 6.2 and Figure 6.2 provide a summary of the blood bacterial count data in theantibacterial clinical trial. A first inspection of the box plots in Figure 6.2 suggests somerather peculiar behavior in the placebo subjects: the distribution of this group appears tohave a very long and significant tail upwards away from zero. Moreover, the data are quiteskewed (as evidenced by the difference between the mean and median in the summarystatistics). Note that despite the rather large difference in the two means, the medians arethe same. A final feature worth examining is the seemingly large difference in dispersionbetween the two groups.

A rather naıve way to approach a comparison of the two location parameters wouldbegin with a simple application of a Student’s two-sample t-test based on the TTESTprocedure (Program 6.3).

Page 137: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

124 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 6.2 Results of the antibacterial clinical trial

Placebo Treated

0

100

200

300

Blo

od b

acte

rial c

ount

Group

Program 6.3 Student’s two-sample t-test in the antibacterial trial example

proc ttest data=bbc;ods select ttests;class group;var bbc;run;

Output from Program 6.3

T-Tests

Variable Method Variances DF t Value Pr > |t|

bbc Pooled Equal 291 5.28 <.0001bbc Satterthwaite Unequal 130 4.40 <.0001

Output 6.3 shows that the two- and one-tailed p-values (assuming unequal variances)are highly significant (p < 0.0001). It will be unwise to be very happy at this statisticallysignificant result (we have evidence that a statistically significant decrease in the treatmentmean exists) and more prudent to reflect on the apparent lack of symmetry in the data.Given the observed asymmetry, one can decide that perhaps an independent two-samplenonparametric test might be more appropriate in this setting. Program 6.4 performs theWilcoxon Rank Sum test on the BBC data set.

Program 6.4 Wilcoxon Rank Sum test in the antibacterial trial example

proc npar1way data=bbc wilcoxon;ods select WilcoxonTest;class group;var bbc;run;

Page 138: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 125

Output from Program 6.4

Wilcoxon Two-Sample Test

Statistic 17223.0000

Normal ApproximationZ 1.7149One-Sided Pr > Z 0.0432Two-Sided Pr > |Z| 0.0864

t ApproximationOne-Sided Pr > Z 0.0437Two-Sided Pr > |Z| 0.0874

Z includes a continuity correction of 0.5.

The one-tailed p-value (based on the normal approximation) displayed in Output 6.4 issignificant (p = 0.0432). Once again, we appear to have a statistically significant difference(this time, it is a difference between the two medians); however, this conclusion appears tobe at odds with the descriptive statistics in Table 6.2 which indicate that the two groupshave a median of 8.0.

The problem with the second analysis, although it accounts for a lack of symmetry inthe two distributions, is that it does not account for a difference in dispersion or scale. Oneof the chief assumptions of the Wilcoxon Rank Sum test is that the two samples have thesame level of dispersion. This feature of our study could be the cause of the seeminglystrange statistical results. It appears that a difference exists in the dispersion of the twodistributions, yet does a more objective analytical means exist to substantiate this belief?

Comparison of Two Distributions’ DispersionOne means to compare the dispersion or spread of two distributions, assuming that themedians are identical, is to use the Ansari-Bradley test (Ansari and Bradley, 1960).Inference about the equality of two distributions’ variation is based upon the ratio of thetwo distributions’ scale parameters. If X comes from a distribution F (μ, ϕ1), with locationparameter μ and scale parameter ϕ1, and Y comes from a distribution F (μ, ϕ2), withlocation parameter μ and scale parameter ϕ2, we are interested in φ = ϕ1/ϕ2. If φ is closeto unity, evidence does not exist to declare the two distributions to have differing amountsof variation. Otherwise, one distribution is declared to have greater dispersion than theother. Note that the Ansari-Bradley test assumes that the two distributions have acommon location parameter, μ.

What if we are working in an area of research where we really do not have knowledgeabout the assumption of a common location parameter, μ? A way around makingassumptions in this setting is to transform each of the n1 X values and the n2 Y values bysubtracting each group’s median from the original raw data:

X∗i = Xi − mX , i = 1, . . . , n1, Y ∗

j = Yj − mY , j = 1, . . . , n2,

where mX and mY are the medians of the X’s and Y ’s, respectively. To construct theAnsari-Bradley statistic, we begin by jointly ordering the X∗’s and Y ∗’s from smallest tolargest. The ranking procedure is slightly different from most nonparametric techniques.Table 6.3 shows how the ranking procedure works for the Ansari-Bradley test. The ranksare assigned from the “outward edges, inward,” for the ordered values. Ties are resolved asbefore in the calculation of the Wilcoxon Rank Sum test (see Section 6.2.2).

Page 139: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

126 Pharmaceutical Statistics Using SAS: A Practical Guide

Table 6.3 Ranking Procedure for the Calculation of the Ansari-Bradley Statistic

Step Procedure

0 Order X∗1 , X∗

2 , . . . , X∗n1

, Y ∗1 , Y ∗

2 , . . . , Y ∗n2

from smallest to largest,label these values Oi for i = 1, . . . , n1 + n2

1 Assign O1 and On1+n2 rank= 12 Assign O2 and On1+n2−1 rank= 23 Assign O3 and On1+n2−2 rank= 3. . . . . .

For the n1 +n2 ranked values, the Ansari-Bradley statistic is calculated by adding up then2 ranks of the Y ∗ values: S =

∑n2j=1 Rj , where Rj is the rank of Y ∗

j in the combined sample.The null hypothesis H0 : φ = 1 is rejected in favor of HA : φ �= 1 at level α = α1 + α2 if

S ≥ cα1 or S ≤ c1−α2 − 1, where cα1 and c1−α2 are quantiles from the null distribution of theAnsari-Bradley statistic with α1 + α2 = α. If n1 = n2, it is reasonable to pickα1 = α2 = α/2. A corresponding large sample approximate test exists as well as correctionsto the test statistic for the presence of ties. The interested reader is encouraged to read adiscussion of these items in Hollander and Wolfe (1999, Chapter 5).

Let’s now return to the antibacterial trial example. It would be interesting to testwhether the scale parameters of the two distributions are different. If evidence exists thatthe dispersion of the two distributions is different, one of the principal assumptions of theWilcoxon Rank Sum test is violated.

The Ansari-Bradley test may be carried out easily with PROC NPAR1WAY(Program 6.5). The first step in performing the version of the Ansari-Bradley testdescribed above is to determine each sample’s median (this is accomplished by using theUNIVARIATE procedure). After that, each value is adjusted by its location (median) andthe Ansari-Bradley statistic is calculated from the adjusted values using PROCNPAR1WAY with the AB option.

Program 6.5 Ansari-Bradley test in the antibacterial trial example

proc univariate data=bbc;class group;var bbc;output out=medianbbc median=median;

proc sort data=bbc;by group;

proc sort data=medianbbc;by group;

data adjust;merge bbc medianbbc;by group;bbc_star=bbc-median;

proc npar1way data=adjust ab;ods select ABAnalysis;class group;var bbc_star;run;

Page 140: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 127

Output from Program 6.5

Ansari-Bradley One-Way Analysis

Chi-Square 9.8686DF 1Pr > Chi-Square 0.0017

Output 6.5 lists the Ansari-Bradley test statistic (S = 9.8686) and associated p-valuebased on a normal approximation (p = 0.0017). Note that the sample size is sufficientlylarge to warrant the use of the large sample approximate test. At the 0.05 level ofstatistical significance, we can declare that a statistically significant difference existsbetween the two groups with respect to the scale parameter or, equivalently, dispersion ofthe blood bacterial count data.

Fligner-Policello Approach to the Behrens-Fisher ProblemIt is clear from Output 6.5 that one of the chief assumptions of the Wilcoxon Rank Sumtest is violated because the two distributions have differing amounts of variation. Does astatistical inferential procedure exist that will compare two group medians in the face ofunequal spread?

Fligner and Policello (Fligner and Policello, 1981) suggested the following test tocompare two location parameters from distributions with differing amounts of variation.The statistic is based upon a quantity called a placement. A placement is the number ofvalues in one sample strictly less than a given value of a second sample. Consider samplesX1, . . . , Xn1 and Y1, . . . , Yn2 . For a value Xi, i = 1, . . . , n1, its corresponding placement, Pi, isthe number of values from Y1, . . . , Yn2 less than Xi. Similarly, for a value Yj , j = 1, . . . , n2,its placement, Qj , is the number of values from the first sample less than Yj . The nextsteps involve calculating quantities similar to those required for a standard Student’stwo-sample t-test. To perform the hypothesis test

H0 : δ = 0 when ϕ1 �= ϕ2 versus HA : δ > 0 when ϕ1 �= ϕ2,

we need to compute the average placements, P = n−11

∑n1i=1 Pi and Q = n−1

2∑n2

j=1 Qj , as wellas the variances:

V1 =n1∑i=1

(Pi − P )2, V2 =n2∑

j=1

(Qj − Q)2.

The Fligner-Policello test statistic is of the form:

F =n1P − n2Q

2√

V1 + V2 + P Q.

We reject H0 in favor of HA at level α if F ≥ cα, where cα is the cutoff from the nulldistribution of the Fligner-Policello statistic. By the Central Limit Theorem, F ∼ N(0, 1),cα may be conveniently replaced with the quantile from a standard normal distribution, zα,when n1 and n2 are large. This gives us the large-sample versions of the lower andtwo-tailed tests, respectively:

• Reject H0 : δ = 0 (when ϕ1 �= ϕ2) in favor of HA : δ < 0 (when ϕ1 �= ϕ2) if F ≤ −zα.• Reject H0 : δ = 0 (when ϕ1 �= ϕ2) in favor of HA : δ �= 0 (when ϕ1 �= ϕ2) if |F | ≥ zα/2.

If ties exist in the data, one needs to adjust the Fligner-Policello statistic defined above.The adjustment occurs at the level of the placements. When calculating one set of the

Page 141: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

128 Pharmaceutical Statistics Using SAS: A Practical Guide

placements, for example, to compute Pi, we count the number of the Y ’s less than Xi andadd half of the values equal to Xi. The same is true for the other set of placements. Thebalance of the method remains unchanged.

%FPSTATISTIC Macro for Performing the Comparison of Two Location Parametersin the Absence of Unequal ScaleThe %FPSTATISTIC macro, available on the book’s companion Web site, performs thelarge sample version of the Fligner-Policello test. To invoke the macro, the user needs tospecify three parameters:

• DATASET is the name of the data set.• VAR is the name of the response variable.• GROUPVAR is the name of the grouping (classification) variable for the macro. This

variable must have only two levels.

The macro begins by determining the sample size for each level of the grouping variable.After the macro determines the two group sample sizes, it begins to calculate theplacements for each group including an adjustment for ties. The algorithm involves a loopthat iteratively compares values and assigns an indicator variable with the values 1 (greaterthan response in other group), 0.5 (tied or equal values) or 0 (for less than response inother group). These indicators are added to form the placements for the first group with n1observations. The same procedure is followed for the n2 placements of the second group.

The final portion of the macro uses the MEANS procedure to calculate the sum of theplacements, mean placements, and corrected sums of squares for the placements for eachlevel of the grouping variable. The PROBNORM function is then used to determine thep-values. The macro outputs three versions of the large sample Fligner-Policello test,two-sided, upper and lower (the results are saved in the FPSTATISTIC data set).

Program 6.6 calls the %FPSTATISTIC macro to perform a comparison of the bloodbacterial count data in the placebo and treated groups. Note that the location parametersfor the two levels of the classification variable are compared in reverse alphanumeric sortorder. Thus, for the BBC data set, the macro will test whether the location parameter inthe treated group is greater than the location parameter in the placebo group. In theantibacterial trial example, we are interested in knowing if the treatment lowered the bloodbacterial count. Thus we need to focus on the one-sided, lower-tailed test.

Program 6.6 Fligner-Policello test in the antibacterial trial example

%fpstatistic(dataset=bbc,var=bbc,groupvar=group);proc print data=fpstatistic noobs label split="*";

format uhat 6.3 p2sided up1sided low1sided 5.3;var n1 n2 uhat p2sided up1sided low1sided;run;

Output from Program 6.6

Sample Samplesize size FP Two-sided Upper-tailed Lower-tailed

group 1 group 2 statistic p-value p-value p-value

109 184 -1.615 0.106 0.947 0.053

Output 6.6 shows that the one-sided, lower-tailed p-value is not significant at a 0.05level (p = 0.053). Thus the Fligner-Policello test fails to reject the null. We conclude that

Page 142: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 129

there is not enough evidence to declare the blood bacterial counts to be statisticallysignificantly different between the two treatment groups. The results of this analysis aremuch more in agreement with the summary statistics presented earlier in Table 6.2.

Role of Data Transformations

It is very common for data analysts to employ basic transformations on data so that thetransformed data will have desirable properties. Two common means of transforming dataare the use of logarithms and rank-transformations. Logarithmic transformations are notalways useful, as zeros or negative values may exist in data sets. Moreover, applying alogarithmic transformation to data, followed by performing a two-sample Student’s t-test,does not always test the desired hypothesis comparing the locations of two samples’distributions (see Zhou et al., 1997, for a discussion of this practice).

Some folklore has evolved among practitioners of statistical methods in the field withrespect to the rank-transformations. A common misconception is that the rank-transformeliminates the need to adjust for unequal dispersion, i.e., the rank-transformation correctsfor the problem of unequal variation. As illustrated previously, the results of performing arank-transformation and a large-sample approximate Wilcoxon Rank Sum test mayproduce a counter-intuitive inference. The author would recommend the application of theFligner-Policello method in an analysis of two independent samples where the distributionsare believed to differ in scale.

Although, on the surface, the analysis of the two independent samples seems trivial, thesetting still requires the care necessary for any data analysis. The features of the data mustbe understood and matched appropriately to the assumptions of the statistical method(s)employed. This is a theme that will be reiterated more times throughout this chapter.

Now that we have examined a few aspects of the two independent samples case, it istime to advance to the setting of k independent samples. The next section will addressseveral nonparametric approaches to analyzing data in this common experimental setting,largely emphasizing examples from drug discovery research.

6.2.4 SummaryThe two-sample problem is not unlike other problems in statistical inference: carefulexamination of the assumptions is required to execute an appropriate analysis.Nonparametric statistics methods afford techniques for handling the comparison of twodistributions’ locations in both the case of similar dispersion (Wilcoxon Rank Sum test)and differing dispersion (Fligner-Policello test). In the case of the former, the correspondinganalysis may be carried out with PROC NPAR1WAY. In the latter case, the analysis maybe executed with a SAS macro created by the author (%FPSTATISTIC macro).

6.3 The One-Way LayoutThe one-way layout is a commonly used experimental design in biological research. Arecent Web search of the archival scientific literature (Science Direct) covering the domainof published pharmacological, toxicological, and pharmaceutical science research from 2000to first quarter 2005 produced 3,671 hits pertaining to the application of a one-way layoutas the means to address a specific scientific hypothesis. The one-way layout is a powerfulmeans to study basic scientific questions because of its simplicity. Recall that in a one-waylayout, experimental units (e.g., subjects) are randomly assigned to two or more treatmentgroups. The treatment groups could consist of two or more distinct treatments or increasingdoses of a single agent (as in the case of later stage drug discovery studies or many standardtoxicology investigations). Each unit receives its treatment and a continuous measurementis collected. Formally, consider a study (drug discovery experiment or clinical trial) with k

Page 143: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

130 Pharmaceutical Statistics Using SAS: A Practical Guide

treatment groups and ni units/subjects in the ith group. Let Xij be the response of the jthexperiment unit (j = 1, . . . , ni) to the ith treatment (i = 1, . . . , k). The total sample size forthe study is N = n1 + . . . + nk. Let τi be the effect of the ith treatment group. Then, we canexpress the relationship between the treatment and response with a simple linear model:

Xij = τi + εij . (6.1)

where εij represents the measurement error associated with the jth experimental unit(subject) assigned to the ith group (or treatment). We will assume that the εij ’s areindependent and identically distributed by virtue of the original random assignment ofunits to treatments.

EXAMPLE: Dopamine ExperimentIn an effort to make the discussion more concrete, consider the following simple experimentas an example (Juneau, 2004). Samples of PC12 cells were randomized to one of threegroups. The PC12 cells were cultured in one of three media: the first medium was infectedwith a particular strain of bacteria postulated to be associated with the development ofParkinson’s disease. The second group used a medium cultured with a second strain ofbacteria. The third group of cells was cultured in a normal uninfected medium. All cellswere incubated for 24 hours and harvested to determine the dopamine concentration. Dueto some unanticipated circumstances, some samples were lost during processing and theresultant sample sizes were unequal at the end of the study. The dopamine concentrationdata from this study are included in the DOPAMINE data set provided on the book’scompanion Web site.

From the formal statement above, Xij would represent the dopamine concentrationresponse of the jth experiment unit (j = 1, . . . , ni) to the ith treatment (i = 1, 2, 3), wheretreatment 1 (control group) sample size is n1 = 14, treatment 2 (Strain I) sample size isn2 = 7, and treatment 3 (Strain II) sample size is n3 = 10.

Typically, researchers will be interested in making decisions about the τi’s: “Doesevidence exist to declare the response of at least one of the treatment groups to be differentfrom the others?” Mathematically, one could ask: “Is it the case that τ1 = τ2 = . . . = τk ordoes evidence exist to declare at least one τi �= τl for i �= l?” For the dopamine experimentillustrated previously, this question would translate into “How does the presence of one ofthe three treatments affect dopamine concentration?” Once again, one could express thisquestion mathematically: “Is it the case that τ1 = τ2 = τ3 or that at least one differenceexists among all pairs of τ1, τ2, or τ3?”

In some sense, evidence to reject the idea that the treatment effects are all the same isnot very informative. Typically, such conclusions do not establish differences in activityamong several novel agents. Investigators will often be more interested in examiningquestions with alternatives designed for the comparison of one or more of the treatmenteffects against a designated group (e.g., a control group) or elucidating where treatmenteffects exist among the k treatments (e.g., pair-wise comparisons). This section will bedevoted to the examination of various alternative hypotheses that arise in typicalpharmaceutical investigations where the one-way layout is the design of choice.

Section 6.3.1 will address the general alternative hypothesis of unequal treatment effectsassociated with the Kruskal-Wallis test. Examples will be demonstrated with PROCNPAR1WAY. Section 6.3.2 will deal with nonparametric multiple comparison proceduresin the one-way layout when an investigator is interested in comparing all treatment groupsagainst a single designated (control) group or performing all simultaneous pair-wisecomparisons of treatment effects. Section 6.3.2 will introduce an original SAS macro codeto perform the desired statistical tests.

Page 144: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 131

6.3.1 Testing a General Alternative for k Groups: The Kruskal-Wallis TestTable 6.4 and Figure 6.3 summarize the dopamine concentration response data collected inthe dopamine experiment. Notice the slight asymmetry in the distribution patterns of thecontrol and Strain I-infected samples. The control group has two responses that seemextreme relative to the rest of the measurements.

Table 6.4 Summary of the Dopamine Concentration Response Data in the Dopamine Experiment

Group Sample size Mean Standard deviation Median Interquartile range

Control 14 100.00 15.14 95.24 13.71Strain I 7 63.12 28.65 54.29 46.38Strain II 10 78.04 29.81 72.98 45.12

Figure 6.3 Results of the dopamine experiment

Control Strain I Strain II

25

50

75

100

125

Dop

amin

e le

vel

Group

A standard approach to comparing these three treatments would be to begin with aclassical linear model relating the dopamine response to the treatment:

Xij = μ + τi + ηij ,

where for i = 1 (controls), j = 1, . . . , 14; i = 2 (Strain I), j = 1, . . . , 7; i = 3 (Strain II),j = 1, . . . , 10, ηij are independent and identically distributed Gaussian errors (N(0, σ2)),and μ represents the overall population dopamine concentration mean response. Thismodel could then be fit with the GLM procedure. Let’s examine a plot of the density of theresiduals after fitting the dopamine concentration data with a standard linear model basedupon analysis of variance assumptions.

The two curves superimposed in Figure 6.4 allow us to make an interesting conclusion.The dashed curve represents a Gaussian (normal) distribution fitted to the residuals. Asolid kernel smooth curve fit to the same residuals suggests that the residuals areasymmetrical (this is also demonstrated by the separation of the means and medians inTable 6.4).

It appears by the evidence presented above that the normality assumption of the ηij’s isin question. The typical parametric model and inferences based upon the analysis ofvariance are thus inappropriate for the evaluation of this experiment. A more appropriate

Page 145: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

132 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 6.4 Plot of residuals after fitting a traditional analysis of variance-type model in the dopamine experiment

Ideal Gaussiandistributedresiduals

Distribution ofresiduals after

kernel-smoothing

Residuals

-40 -20 0 20 40 60

analysis would be based upon the model introduced earlier in this section and theKruskal-Wallis test (Kruskal and Wallis, 1952).

Consider the model introduced at the beginning of Section 6.3.1 that relates theresponse of an experimental unit to the effect of the ith treatment τi. The Kruskal-Wallistest is used to test the following null hypothesis against a very general alternative:

H0 : τ1 = τ2 = · · · = τk

versus

HA : at least one τi �= τl for1 ≤ i ≤ k, 1 ≤ l ≤ k, i �= l.

In the dopamine study, H0 : τ1 = τ2 = τ3 is tested versus the alternative that states that atleast two out of three τ ’s are different from each other.

The test statistic for the Kruskal-Wallis test is constructed by ranking all Nobservations from the smallest value to the largest. Tied values are assigned the averagerank. The mean rank is then calculated for each group. Denote the mean rank for the ithgroup by Ri. The Kruskal-Wallis test statistic, K, is then defined by

K =12T

N(N + 1)

k∑i=1

ni

(Ri − N + 1

2

)2

,

where T is a correction factor for tied values. If for all N values, ξ groups of size tv(1, 2, . . . , v, . . . , ξ) exist, the correction factor T is expressed as (Hollander and Wolfe, 1999)

T = 1 − 1N3 − N

ξ∑v=1

(t3v − tv).

H0 is rejected in favor of HA at level α, if K ≥ cα.The cutoff value for the test, cα, comes in two varieties. The first is the large sample

approximate test, which is based upon a chi-squared cutoff. The degrees of freedom for thistest are k − 1, i.e., χ2

k−1(α). The other type of cutoff for this test is the exact version, basedupon the permutation distribution of the ranks. PROC NPAR1WAY has had the capability

Page 146: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 133

to perform the exact version of the test since SAS Version 6 via the EXACT statement.The formulation of the Kruskal-Wallis statistic and test stated above is not truly an exactα-level test, but only an approximate α-level test. Fortunately, if ties exist in the data,PROC NPAR1WAY will perform the exact α-level test because it determines thepermutation distribution of the tied ranks when it determines the p-value for the analysis.

Large-Sample Kruskal-Wallis TestProgram 6.7 performs the large-sample Kruskal-Wallis test in the dopamine experimentusing PROC NPAR1WAY. The WILCOXON option is used in the procedure to specifyWilcoxon-type scores to perform the Kruskal-Wallis test.

Program 6.7 Large-sample Kruskal-Wallis test in the dopamine experiment

proc npar1way data=dopamine wilcoxon;class group;var dopamine;run;

Output from Program 6.7

Kruskal-Wallis Test

Chi-Square 9.0710DF 2Pr > Chi-Square 0.0107

Output 6.7 shows that the p-value produced by the large-sample Kruskal-Wallis test is0.0107 and thus we would reject the null hypothesis of equal dopamine response for allthree groups in favor of the alternative that at least one difference exists among the threesets of treated samples at α = 0.05.

Exact Kruskal-Wallis Test

If we examine the DOPAMINE data set, we notice that several tied responses exist for thedopamine response. Tied values are a reality in many real experiments because oflimitations on measurement precision, rounding off of values by scientists, or various otherreasons. Let’s take advantage of the EXACT statement in PROC NPAR1WAY and see ifthe results change dramatically (Program 6.8). The program is virtually identical toProgram 6.7, with the exception of the insertion of the EXACT statement with aWILCOXON option.

Program 6.8 Exact Kruskal-Wallis test in the dopamine experiment

proc npar1way data=dopamine wilcoxon;class group;var dopamine;exact wilcoxon;run;

Page 147: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

134 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 6.8

Kruskal-Wallis Test

Chi-Square 9.0710DF 2Asymptotic Pr > Chi-Square 0.0107Exact Pr >= Chi-Square 0.0074

Output 6.8 lists the exact p-value (p = 0.0074), which is also significant at a 0.05 level.It is important to mention that running the exact Kruskal-Wallis test is veryresource-intensive. The real time run for this short program and relatively small data setwas about three hours long. From the author’s perspective, the additional resourcesrequired may not justify the implementation of the exact test in this particular setting.This may not always be the case, however. Let’s consider another experiment.

EXAMPLE: Hyperplastic Alveolar Nodule (HAN) StudySamples of four types of cell subsets are prepared for a study of the progression of mousemammary preneoplastic hyperplastic alveolar nodule (HAN) line C4 to carcinoma. Hoechstfluorescence is measured as a response for each sample. The data from this study areincluded in the HAN data set given on the book’s companion Web site.

Let’s assume that the small sample size for this study is enough to warrant the selectionof a nonparametric statistical method (see Section 6.1). We also assume that theinvestigator has a very wide alternative of interest: the investigator is interested in evidenceof any difference in response. Does the choice of an exact test or asymptotic test matter?Program 6.9 carries out the large-sample and exact Kruskal-Wallis tests in the HAN study.

Program 6.9 Large-sample and exact Kruskal-Wallis tests in the HAN study

proc npar1way data=han wilcoxon;class celltype;var fluor;exact wilcoxon;run;

Output from Program 6.9

Kruskal-Wallis Test

Chi-Square 6.7315DF 3Asymptotic Pr > Chi-Square 0.0810Exact Pr >= Chi-Square 0.0383

Output 6.9 lists the large-sample and exact p-values produced by the Kruskal-Wallistests. It appears that if the investigator were interested in using the traditional 0.05 level ofstatistical significance, he or she might be disappointed with the results of the asymptotictest (p = 0.0810 > 0.05), or elated at the conclusion reached by the exact Kruskal-Wallisanalysis (p = 0.0383 < 0.05). The point of this illustration is that small sample sizes caninfluence the results of a testing procedure and should be considered before an analysisstrategy is selected. The next section offers some rules of thumb about group size andabout when the asymptotic procedures typically perform reasonably.

Page 148: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 135

6.3.2 Nonparametric Multiple Comparison Procedures for Specific AlternativesA common objective of many pharmaceutical research investigations with a one-way layoutis to compare treatments in a pair-wise fashion. Investigators are typically interested incomparing all treatment responses against one another or in comparing several agentsagainst a designated group (e.g., an inactive control group). Both of these settings involvethe simultaneous comparison of several group location parameters (say, means in theparametric case, medians in the nonparametric). It is also often desirable to preserve thefamily-wise error rate in this inference; i.e., the probability of declaring at least onetreatment to be different from another, when in fact, it is not.

If we return to the dopamine experiment first presented in Section 6.3.1, we can see anexample of a discovery biology study where investigators might be interested in performingall pair-wise comparisons of treatment groups. The investigators would certainly beinterested in comparing the typical response in the Strain I and Strain II infected sampleswith that of the untreated media samples (control); however, it might be of scientificconsequence to know if a difference exists between the two sets of infected samples as well.

Dunn’s Procedure for All Pair-Wise Comparisons

If an investigator is interested in comparing the location parameters of k experimentalgroups simultaneously and preserving the family-wise error rate, he or she could use anapproach suggested by Dunn (Dunn, 1964) for the linear model introduced previously(6.1), i.e., conclude that τi �= τl if

|Ri − Rl| > zα/k(k−1)

√N(N + 1)

12

(1ni

+1nl

),

where Ri is the mean of the joint ranks for the ith group, Rc is the mean of the joint ranksfor the lth group, and ni and nl are sample sizes in the two groups, respectively, N is thetotal sample size, k is the total number of groups (in the dopamine experiment, k = 3) andzα/k(k−1) is the 100α/[k(k − 1)]th upper quantile from a standard Gaussian distribution.

Recall that the joint ranking of a data set is determined by ranking all of the Nobservations together from smallest to largest.

Dunn’s procedure offers the following advantages:

• The symmetry assumption, which is often difficult to assess in drug discovery settingswith small sample sizes, may be relaxed or ignored.

• Equal sample sizes are not required.• Relatively small total sample sizes may be analyzed with this technique, i.e., three

groups with five experimental units/group or more than three groups with fourunits/group (see Lehman, 1975).

%DUNN MacroA simple set of macros can be constructed in SAS to perform Dunn’s procedure for allpair-wise comparisons. The author composed a simple SAS macro to perform Dunn’sprocedure as part of a presentation for the 2004 PharmaSUG meeting in San Diego(Juneau, 2004). The %DUNN macro was designed to imitate PROC NPAR1WAY, withrespect to ease of use and input of information, and to produce output quite similar toSAS’ very popular procedure, GLM, using the MEANS statement and the CLDIFF option.The macro can be found on the book’s companion Web site.

The %DUNN macro consists of a body of code containing one embedded macro(%GROUPS). The embedded macro determines the number of groups present and assignsthat value to a macro variable (NGRPS). If a group in the data set does not contain at

Page 149: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

136 Pharmaceutical Statistics Using SAS: A Practical Guide

least one response value (i.e., all values are missing), it will not be included in the analysis.The embedded macro also creates one global macro variable that contains the group labels(GRPVEC) for the levels of the class variable. The main body of the SAS code determinessummary statistics (e.g., average ranks, sample sizes, etc.). This information is employed tocalculate the pair-wise test statistics. The corresponding cutoff for the test statistic iscalculated with the PROBIT function. Results are then printed out using the PRINTprocedure.

Program 6.10 carries out Dunn’s test in the dopamine experiment by invoking the%DUNN macro. The first parameter of the macro is the input SAS data set name(DOPAMINE), the second the classification or grouping variable (GROUP), the third thename of the response variable in the input data set (DOPAMINE), and the fourth macroparameter is the overall family-wise error rate (0.05).

Program 6.10 Large-sample and exact Kruskal-Wallis tests in the HAN study

%dunn(dopamine,group,dopamine,0.05);

Output from Program 6.10

Large sample approximation multiple comparison proceduredesigned for unbalanced data

3 groups: Control StrainI StrainII (respective sample sizes: 14 7 10)Alpha = 0.05

Differencein Cutoff

Comparison Group average at Significancenumber comparisons ranks alpha=0.05 difference = **

1 Control-StrainI 11.8571 10.0759 **2 Control-StrainI 7.6429 9.01213 StrainI-StrainI 4.2143 10.7266

Output 6.10 displays the output of the %DUNN macro. The %DUNN macro producesTITLE statements that state the number of class levels, a list of each of the class levelswith non-missing values, and the corresponding group sample sizes. Moreover, the macrogenerates output that contains the relevant test statistic for each comparison (a function ofthe average ranks/class level), the corresponding cutoff for the chosen level of family-wiseerror, and a symbol indicating whether the results of the statistical inference arestatistically significant. From this analysis, one would conclude that a statisticallysignificant difference existed between the median dopamine levels in the controls andsamples treated with the first strain of bacteria.

Dwass-Steel-Critchlow-Fligner Procedure for All Pair-Wise ComparisonsThe Dwass-Steel-Critchlow-Fligner (DSCF) procedure is another popular form ofsimultaneous nonparametric inference in the one-way layout for all pair-wise comparisons(Hollander and Wolfe, 1999). If a pharmaceutical researcher is interested in comparing thelocation parameters of k experimental groups (τ1, . . . , τk) simultaneously, and preservingthe family-wise error rate, he or she could use an approach suggested by Dwass (Dwass,1960) and Steel (Steel, 1960) for the linear model previously introduced (6.1).

The procedure begins by calculating the k(k − 1)/2 pairs of Wilcoxon Rank Sumstatistics, Wil (Wilcoxon, 1945) for each pair, i and l (1 ≤ i ≤ l ≤ k). The Wilcoxonstatistics should include an adjustment for tied values. Conclude that τi �= τl if |Dil| > qα,

Page 150: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 137

where

Dil =

√2

Var(Wil)

(Wil − min(ni, nl)(ni + nl + 1)

2

),

Var(Wil) is the tie-adjusted variance for the Wilcoxon statistic, and qα is the 100αth upperquantile from the Studentized Range distribution (Harter, 1960).

Suppose that for all ni + nl observations in the comparison, ξ tied groups of size tv(1, 2, . . . , v, . . . , ξ) exist. The variance in the denominator of the statistic is given by(Hollander and Wolfe, 1999)

ninl

24

(ni + nl + 1 −

∑ξv=1(tv − 1)tv(tv + 1)

(ni + nl)(ni + nl − 1)

).

%DSCF Macro

An analysis using the DSCF method may be conducted using a SAS macro called %DSCFcreated by the author for a 2004 PharmaSUG presentation (Juneau, 2004). The call of the%DSCF macro is as follows:

%dscf(dataset,group,response,alpha);

The first parameter of the macro is the input data set name (DATASET), the secondthe classification or grouping variable (GROUP), the third the name of the responsevariable in the input data set (RESPONSE), and the fourth macro parameter is the overallfamily-wise error rate (ALPHA).

The %DSCF macro consists of a body of code containing one embedded macro(%GROUPS). The embedded macro determines the number of groups present (NGRPS) asin the %DUNN macro. If a group in the input data set does not contain at least oneresponse value, it will be excluded from the analysis. The embedded macro also creates twoglobal macro variables that contain the group labels (GRPVEC) for the levels of the classvariable and information about the sample size for each group (NVEC). The main body ofthe code calculates the necessary summary statistics (e.g., Wilcoxon Rank Sum teststatistics) and the number of ties present in each pair-wise comparison. This information isthen used to calculate the pair-wise test statistics. The cutoff for the test statistic iscalculated with the PROBMC function (using the Studentized Range distribution). As themacro iterates between all pair-wise comparisons it concatenates successive results in adata set called STAT. The final results are then printed out with PROC PRINT.

EXAMPLE: Cardiovascular Discovery StudySubjects were randomized to one of four treatment groups: three active agents and onevehicle control group. The goal of the experiment was to determine whether the agentscould affect triglyceride level (in mg/dl) relative to the vehicle controls and to determinewhether evidence existed to declare one agent different from another with respect totriglyceride response. Each subject was treated and, after a fixed post-treatment period,the blood triglyceride level was measured for each subject.

The data gathered in the study are included in the TRIG data set provided on thebook’s companion Web site. The results of the experiment are displayed in Table 6.5 andFigure 6.5 with box plots and summary statistics. Note the balanced sample sizes. TheDSCF multiple comparison method works optimally under settings with equal sample sizesper group.

The triglyceride data can be analyzed using the %DSCF macro designed to perform thedesired simultaneous pair-wise nonparametric comparisons of all treatments (seeProgram 6.11).

Page 151: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

138 Pharmaceutical Statistics Using SAS: A Practical Guide

Table 6.5 Summary of Triglyceride Data in the Cardiovascular Discovery Study

Group Sample size Mean Standard deviation Median Interquartile range

Vehicle 5 99.8 16.71 105.0 20.00Treatment A 5 78.2 29.75 71.0 26.00Treatment B 5 49.0 17.33 42.0 24.00Treatment C 5 59.0 15.00 62.0 26.00

Figure 6.5 Results of the cardiovascular discovery study

Trt A Trt B Trt C Vehicle

25

50

75

100

125

Trig

lyce

ride

leve

l

Group

Program 6.11 Dwass-Steel-Critchlow-Fligner (DSCF) method in the cardiovascular discovery study

%dscf(trig,group,trig,0.05);

Output from Program 6.11

Large sample approximation multiple comparison procedure

or all treatment pairsbased upon pairwise rankings

4 groups: TreatmentA TreatmentB TreatmentC Vehicle (respective sample sizes: 5 5 5 5)

Alpha = 0.05

Test statistic Cutoff

Group Comparison Test absolute at Significantcomparisons number statistic value alpha=0.05 difference = **

TreatmentA - TreatmentB 1 -2.21565 2.21565 3.63316TreatmentA - TreatmentC 2 -1.62481 1.62481 3.63316TreatmentA - Vehicle 3 1.92023 1.92023 3.63316TreatmentB - TreatmentC 4 1.92023 1.92023 3.63316

TreatmentB - Vehicle 5 3.69274 3.69274 3.63316 **TreatmentC - Vehicle 6 3.69274 3.69274 3.63316 **

The output of the %DSCF macro in Output 6.11 includes TITLE statements that statethe number of class levels, a list of each of the class levels with non-missing values and thecorresponding group sample sizes. The macro also lists the relevant test statistic for eachcomparison (a function of the Wilcoxon statistic), the corresponding cutoff for the chosen

Page 152: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 139

level of family-wise error, and a symbol indicating whether the results of the statisticalinference are statistically significant. We conclude from Output 6.11 that a statisticallysignificant difference existed between the median triglyceride levels in the controls andsamples treated with Treatments B and C. Statistically significant differences did not existamongst the three active agents (Treatments A, B, and C).

Comparison of Dunn and Dwass-Steel-Critchlow-Fligner Methods

Is the DSCF method really necessary, when the multiple comparison procedure suggestedby Dunn is a more general case (unequal sample sizes)? The triglyceride data from thecardiovascular discovery study used previously as an example for the application of theDSCF method are analyzed in Program 6.12 with Dunn’s method for all pair-wisecomparisons.

Program 6.12 Dunn method in the cardiovascular discovery study

%dunn(trig,group,trig,0.05);

Output from Program 6.12

Large sample approximation multiple comparison proceduredesigned for unbalanced data

4 groups: TreatmentA TreatmentB TreatmentC Vehicle (respective sample sizes: 5 5 5 5)Alpha = 0.05

Differencein Cutoff

Comparison Group average at Significancenumber comparisons ranks alpha=0.05 difference = **

1 TreatmentA-TreatmentB 6.6 9.871452 TreatmentA-TreatmentC 3.6 9.871453 TreatmentA-Vehicle 5.0 9.871454 TreatmentB-TreatmentC 3.0 9.871455 TreatmentB-Vehicle 11.6 9.87145 **6 TreatmentC-Vehicle 8.6 9.87145

It is instructive to compare Output 6.11 (DSCF method) and Output 6.12 (Dunnmethod). Recall that in Output 6.11, Treatment C was also declared to be statisticallysignificantly different from the vehicle control group for the median triglyceride response. Acomparison of the properties of these two methods may shed some light on the reason forthe differing inferential conclusions. First, the Dunn method uses a Bonferroni-likecorrection to the family-wise error rate (Miller, 1981) and might be a bit too conservative.Second, the Dunn method employs joint ranking, and thus the comparison of two groups ishighly influenced by the behavior of other groups in the experiment as the data are initiallyranked over the entire experiment (Hollander and Wolfe, 1999). The balanced sample sizesfor all groups also suggest that the DSCF method might be the most appropriate techniqueto employ for all pair-wise nonparametric comparisons.

Simultaneous Nonparametric Inference in the One-Way Layout for All Group Comparisonswith a Designated Control GroupIt is often the case that investigators are interested in comparing the location parametersof treatments against the location parameter of a designated group. This designated groupcould be a control group consisting of the response of subjects or units completely

Page 153: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

140 Pharmaceutical Statistics Using SAS: A Practical Guide

untreated by any form of active agent. These are often referred to as untreated controlsubjects or units. More commonly, a group of experimental units or subjects treated withthe vehicle, or medium for delivery of the compound, is the designated group of interest.Such subjects or units are referred to as forming the vehicle control group. A third type ofcontrol group is a set of subjects or units treated with an agent known to provoke aresponse that is the same as one desired by proponents of one of the compounds underexamination. Such a collection of subjects or units is called a positive control group.

Other forms of designated control groups exist. The point, however, is that ifpharmaceutical researchers are interested in comparing the location parameters of severalexperimental groups (τ1, . . . , τk) to a designated group (τc), simultaneously and preservingthe family-wise error rate, they can use one of two approaches originally suggested byDunn (Dunn, 1964) or Miller (Miller, 1966) for the linear model (6.1). These approachesare described below.

Dunn’s Method for All Group Comparisons to a Designated Control Group(Unequal Sample Sizes)Using the notation first introduced in the beginning of Section 6.3, suppose that ni �= nj forat least one (i, j) pair (1 ≤ i < j ≤ k) of treatments. We could conclude that τi �= τc if

|Ri − Rc| > zα/2(k−1)

√N(N + 1)

12

(1ni

+1nc

),

where Ri is the mean of the joint ranks for the group i, Rc is the mean of the joint ranksfor the control group c, and ni and nc are sample sizes for group i and the control group, c,respectively, N is the total sample size, k − 1 is the total number of comparisons desired,and zα/2(k−1) is the 100α/[2(k − 1)]th upper quantile from a standard Gaussian distribution.

Miller’s Method for All Group Comparisons to a Designated Control Group (Equal SampleSizes)Using the notation introduced above, assume that ni = nj for all (i, j) pairs (1 ≤ i < j ≤ k)of treatments. Then τi is declared different from τc if

|Ri − Rc| > |Mα,k−1,∞|

√N(N + 1)

12

(1ni

+1nc

),

where Ri, Rc, ni, nc, N , K are defined as above and |Mα,k−1,∞| is the αth quantile from theStudentized Maximum Modulus distribution (Pillai and Ramachandran, 1954).

%NPARMCC Macro

All nonparametric pair-wise comparisons to a designated control group in a one-way layoutcan be performed with the %NPARMCC macro that can be found on the book’scompanion Web site. The macro consists of a body of code containing one embedded macro(%GROUPS). The embedded macro determines the number of groups present (NGRPS) asin the %DUNN and %DSCF macros. If a group in the SAS data set does not contain atleast one response value, it will be excluded from the analysis. The embedded macro alsocreates one global macro variable that contains the group labels (GRPVEC) for the levelsof the class variable. The main body of the macro determines the necessary summarystatistics (e.g., average ranks, sample sizes, etc.). This information is then employed tocalculate the pair-wise test statistics. The cutoff for the test statistic is calculated with thePROBIT function for Dunn’s method and the PROBMC function (with the StudentizedMaximum Modulus distribution) for Miller’s method. The results are then printed out withPROC PRINT.

Page 154: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 141

EXAMPLE: Thrombosis ExperimentAn experiment was designed to study the effect of increasing the dose of a novel agent onactivated clotting time (ACT). Subjects were randomized to one of four groups: a vehiclecontrol group, a low dose group, a medium dose group, and a high dose group. About 200minutes after receiving treatment, the ACT for each subject was measured (in seconds).The data from this experiment are contained in the THROMB data set (available on thebook’s companion Web site) and its results are shown in Table 6.6 and Figure 6.6 (asbefore, with box plots and summary statistics).

Table 6.6 Summary of Activated Clotting Times in the Thrombosis Experiment

Group Sample size Mean Standard deviation Median Interquartile range

Control 5 64.2 8.11 63.0 6.00Low dose 5 98.4 17.47 106.0 14.00Middle dose 5 91.8 14.06 98.0 16.00High dose 5 156.4 9.40 153.0 2.00

Figure 6.6 Results of the thrombosis experiment

Control Low dose Medium dose High dose

50

75

100

125

150

Act

ivat

ed c

lotti

ng ti

mes

Group

The thrombosis experiment data can be analyzed using the %NPARMCC macrodesigned to perform the desired simultaneous pair-wise nonparametric comparisons of allgroups against a designated control group. Program 6.13 applies the %NPARMCC macroto the THROMB data.

Program 6.13 Dunn’s and Miller’s methods in the thrombosis experiment

%nparmcc(thromb,group,Control,act,0.05);

Page 155: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

142 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 6.13

Large sample approximation multiple comparison procedurefor all treatment compared against a control (control group = Control)

4 groups: Control HighDose LowDose MedDose (respective sample sizes: 5 5 5 5)Alpha = 0.05

Difference Dunn Millerin cutoff cutoff Significant Significant

Group Comparison average at at difference difference

comparisons number ranks alpha=0.05 alpha=0.05 Dunn’s = *D* Miller’s = *D*

HighDose-Control 1 14.6 8.95745 8.93410 *D* *M*LowDose-Control 2 7.6 8.95745 8.93410

MedDose-Control 3 6.2 8.95745 8.93410

The output of the %NPARMCC macro displayed in Output 6.13 includes a list of theclass levels with non-missing values, corresponding group sample sizes, and relevant teststatistics for each comparison (the difference in the mean ranks), the corresponding cutofffor the chosen level of family-wise error, and a symbol indicating whether the results of thestatistical inference are statistically significant by Dunn’s method (*D*) and Miller’smethod (*M*). We conclude from Output 6.13 that a statistically significant differenceexisted between the median activated clotting time in the controls and samples treatedwith a high dose of the new agent. As the experiment consisted of balanced sample sizes,the conclusion presented by the Miller approach would be considered the appropriate oneto report.

Comparison of Dunn’s and Miller’s Methods for All Pair-Wise Comparisons Versus aDesignated Control GroupIt is interesting to compare the behavior of Dunn’s and Miller’s respective methods using adata set that is not balanced with respect to sample size. Suppose that we conducted asimilar thrombosis experiment as described previously a second time, yet this time withunequal sample sizes. The experimental results are slightly different (the control and lowdose groups have different sample sizes than in the original thrombosis experiment). Theresults are summarized in Table 6.7 and Figure 6.7. The data from this study are includedin the THROMB2 data set available on the book’s companion Web site.

Table 6.7 Summary of Activated Clotting Times in the Thrombosis Experiment with Unequal Sample Sizes

Group Sample size Mean Standard deviation Median Interquartile range

Control 9 62.2 11.10 63.0 6.00Low dose 8 100.0 20.87 98.5 26.00Middle dose 5 96.4 18.37 106.0 24.00High dose 5 144.4 31.63 153.0 2.00

The data from the thrombosis experiment with unequal sample sizes were once againanalyzed using the %NPARMCC macro (Program 6.14).

Program 6.14 Dunn’s and Miller’s methods in the thrombosis experiment with unequal sample sizes

%nparmcc(thromb2,group,Control,act,0.05);

Page 156: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 143

Figure 6.7 Results of the thrombosis experiment with unequal sample sizes

Control Low dose Medium dose High dose

50

75

100

125

150A

ctiv

ated

clo

tting

tim

es

Group

Output from Program 6.14

Large sample approximation multiple comparison procedurefor all treatment compared against a control (control group = Control)

4 groups: Control HighDose LowDose MedDose (respective sample sizes: 9 5 8 5)

Alpha = 0.05

Difference Dunn Millerin cutoff cutoff Significant Significant

Group Comparison average at at difference differencecomparisons number ranks alpha=0.05 alpha=0.05 Dunn’s = *D* Miller’s = *D*

HighDose-Control 1 17.4667 9.42126 10.5710 *D* *M*LowDose-Control 2 10.2917 8.20747 9.2091 *D* *M*MedDose-Control 3 10.1667 9.42126 10.5710 *D*

Output 6.14 displays the cutoffs computed by the %NPARMCC macro in the modifiedthrombosis experiment data set. The results of the analysis are quite interesting. Note thatMiller’s method does not declare the median response for the medium dose group to bestatistically significantly different from the control group, while Dunn’s method doesdeclare this result to be statistically significant. The difference in the inferential conclusionscan be attributed to the fact that Dunn’s method was derived to handle the unequal samplesize setting, whereas the Miller method, using the Studentized Maximum Modulus cutoff,works optimally under conditions of equal sample size (Hochberg and Tamhane, 1987).

Summarization of the Nonparametric Multiple Comparison MethodsTable 6.8 summarizes the nonparametric methods presented in this section and providesadvice on the method to use given the study design of a particular investigation.

Page 157: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

144 Pharmaceutical Statistics Using SAS: A Practical Guide

Table 6.8 A Summary of the Nonparametric Multiple Comparison Methods Discussed in Section 6.4

Desired Study design Nonparametric SAS macro tocomparison feature method perform analysis

All pair-wise Equal group Dwass-Steel- %DSCFcomparison sample sizes Critchlow-Fligner

methodUnequal group Dunn’s method %Dunnsample sizes

Comparisonswith a designated

control group

Equal group Miller’s %NPARMCCsample sizes method

Unequal group Dunn’s method %NPARMCCsample sizes

6.3.3 SummaryThe one-way layout is commonly used in scientific investigations in the pharmaceuticalindustry. For asymmetrical response data, nonparametric statistical methods offer dataanalysis methods for a general alternatives of at least one change in location(Kruskal-Wallis test) or more specific, such as all pair-wise comparisons of treatment grouplocations (Dunn and Dwass-Steel-Critchlow-Fligner methods) or comparisons with adesignated control group (Dunn and Miller methods). All techniques can be useful insettings where a small number of responses in a group skew responses or adversely affect themean response. As was the case for the two-sample setting, it is important to study dataproperties (i.e., balanced vs. unbalanced sample sizes) before selecting a statistical method.

6.4 Power Determination in a Purely Nonparametric SenseThe title of this section must seem a bit of an enigma. What does the expression “in apurely nonparametric sense” really mean? Let’s begin by reviewing some basic concepts inexperimental design and power in the familiar parametric sense. We will then demonstratea nonparametric approach to the same setting. Lastly, we will compare and contrast thetwo approaches to highlight important features.

Consider the design of a simple two-group experiment. Subjects or samples arerandomized to one of two groups and receive a treatment. After treatment, a measurementis collected and the investigators wish to see if evidence exists to declare the responses ofthe two groups to be “different”.

What does it mean for two treatments to be different? Typically, the investigators areinterested in finding a statistically significant effect at some level (say, 0.05) with somereasonable degree of certainty (say, at least 80% of the time). Using some commonsymbolism from mathematical statistics, the latter expression may be written as:

Test H0 : μ1 − μ2 = 0 vesus HA : μ1 − μ2 = δ at some level α,

where:

• α = P (Conclude μ1 − μ2 > 0|(μ1 − μ2) = 0),• 1 − β = P (Conclude μ1 − μ2 > 0|(μ1 − μ2) = δ).

In other words, we are interested in finding a statistically significant difference at an α levelwith 1 − β power when the true difference is δ. If we make the standard assumptions of twoindependent and identically distributed Gaussian samples (N(μ1, σ

2) and N(μ2, σ2),

Page 158: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 145

respectively), we can perform such a comparison via a two-sample Student’s t-test, i.e.,reject H0 in favor of HA if

t =X1 − X2√

2s2/n> T2(n−1)(α),

where X1 and X2 correspond to the sample means of Treatment Groups 1 and 2,respectively, s2 is the pooled variance estimator, and T2(n−1)(α) is a quantile from astandard Student’s t-distribution with 2(n − 1) degrees of freedom.

Naturally, the investigators are interested in the total number of experimental units orsubjects, 2n, required to conduct the experiment. The sample size per group, n, may bedetermined iteratively, using a standard inequality:

n ≥2s2(T2(n−1)(α) + T2(n−1)(β))2

δ2 ,

where T2(n−1)(α) and T2(n−1)(β) are quantiles from a standard Student’s t-distribution with2(n − 1) degrees of freedom.

Note the key features of this method of sample size determination are

• The person using this method needs to feel fairly confident that his or her measurecomes from a distribution that is Gaussian (normal).

• He or she must have a reasonable idea of what the treatment effect should be.• He or she must also have a reasonable estimate of the spread or dispersion of the desired

effect (s2).

It is generally difficult for scientific investigators (based on the author’s personalexperience in statistical consulting) to provide information about a measurement’sdistribution. In fact, it is sometimes very difficult in some scientific disciplines (e.g.,toxicology) for investigators to predict the size of effect that will be elicited by a treatmenta priori.

One possible way to solve this problem is to conduct a small pilot study to get someinitial intuition about a measurement’s properties. Such a study will provide investigatorswith insight into the size of treatment effect that is achievable, as well as some preliminaryideas about the measurement’s properties, such as its pattern of dispersion. However, willsuch a study address the fundamental question of distribution (e.g., Gaussian distribution)when the sample size is small?

A distribution-free, or nonparametric, approach to sample size estimation frees scientistsfrom guessing about the distribution of measurement or treatment effect. One common wayto estimate a sample size in a study with two independent groups, suggested by Collingsand Hamilton (1988), requires that investigators perform a small pilot study. Investigatorsthen use the pilot study’s results to refine their estimates of achievable treatment effect.The scientists must also provide an estimate of their desired statistical significance level;however, they do not have to have any estimate of the dispersion or variation of themeasurement at the time of the sample size determination. This information can beindirectly gleaned from the pilot study. For a given pair of sample sizes, say, n1 and n2,scientists determine the level of power associated with the comparison.

6.4.1 Power Calculations in a Two-Sample CaseHow does the Collings-Hamilton technique work? Suppose the pilot study consists of datafrom two groups, say, X1, . . . , Xn1 and Y1, . . . , Yn2 . For the moment, let’s assume thatn1 = n2 = n and thus the total pilot sample size is 2n observations. Suppose we areinterested in determining the power of a comparison of two treatment groups’ location

Page 159: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

146 Pharmaceutical Statistics Using SAS: A Practical Guide

parameters, one shifted by an amount δ from the other, at the α-level of statisticalsignificance, with a total sample size of 2m (m samples per treatment group). We begin bymaking a random selection (with replacement) of size 2m from the pilot study data, i.e.,X1, . . . , Xn1 , Y1, . . . , Yn2 . Denote the first m randomly sampled observations by R1, . . . , Rm.For the second set of randomly selected observations (denoted by Rm+1, . . . , R2m), add thechange in location, δ, to each of the m values. Denote the resulting valuesRm+1 + δ, . . . , R2m + δ by S1, . . . , Sm. Now compare R1, . . . , Rm with S1, . . . , Sm via a form oftwo-sample location comparison test, with a statistic, say, W , at level α. Create anindicator variable, I, and let I = 1 if W ≥ cα (cα represents the α-level cutoff of the test)and 0 otherwise.

We can repeat the process B times (for some large value of B, i.e., B > 1000), recordingthe value of the indicator variable I as Ib for the bth repetition of the process(b = 1, . . . , B). An estimate of the power, π(α, β,m), of the test conducted at the α-level ofsignificance, for shift of size and sample size m per group is

π(α, β,m) =1B

B∑b=1

Ib.

Collings and Hamilton went on to show in their paper that the described procedure canbe improved in the following way. Consider X1, . . . , Xn and randomly sample from thegroup 2m times. For the second half of the 2m terms, add the shift value δ to each term:Xm+1 + δ, . . . , X2m + δ. Perform the pair-wise comparison at level α between X1, . . . , Xm

and Xm+1 + δ, . . . , X2m + δ, record the result via an indicator variable, and repeat theprocedure B times, as described previously. The same procedure is carried out forY1, . . . , Yn. Let πX(α, β,m) be the power calculated for the Xi’s and πY (α, β,m) be thepower calculated for the Yi’s. The power for the comparison of X1, . . . , Xn and Y1, . . . , Yn

with respect to a shift δ, at level α, for total sample size 2m is:

π(α, β,m) =12(πX(α, β,m) + πY (α, β,m)).

Earlier in our discussion we assumed n1 = n2 = n. This condition is not necessary. Ifn1 �= n2, this approach can be modified by using a weighted mean of the two power values;see Collings and Hamilton (1988) for details.

6.4.2 Power Calculations in a k-Sample CaseMahoney and Magel (1996) extended the results of Collings and Hamilton to cover the caseof k independent samples and their comparison by the Kruskal-Wallis test. The procedureis similar in spirit to that described previously. Consider the testing problem defined inSection 6.3, i.e., consider a study with k treatment groups and ni units/subjects in the ithgroup. In the two-sample case, we talk about a single shift in location, δ. For a k-sampleone-way layout, we have to talk about a k-dimensional vector, δ = (δ1, . . . , δk), where δi isthe shift-change in location of the ith group. Without loss of generality, we assume that thefirst delta component is zero, i.e., δ1 = 0. As was the case in the two-sample setting, weagain need to randomly sample (with replacement) from each of the groups. For the ithgroup, we sample km observations. We then apply the k location shifts to each of the kmobservations as follows. The first m observations are unshifted because δ1 = 0. The secondset of m observations are then shifted by δ2, the third set by δ3, etc. Mahoney and Magelpoint out that, essentially, we now have k random samples of size m, only differing by ashift in location and thus we can perform a Kruskal-Wallis test at level α. If the test isstatistically significant, we let I1 = 1, otherwise I1 = 0. We repeat the process B times, fora large value of B, recording the value of Ib for the bth repetition of the process. The powerfor the ith group is calculated as before, i.e., πi(α, β,m) = B−1 ∑B

b=1 Ib. The outlined

Page 160: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 147

procedure is repeated for the remaining k − 1 groups and the power for the Kruskal-Wallistest is then simply the average power determined k times:

π(α, β,m) =1k

k∑i=1

πi(α, β,m).

6.4.3 Comparison of Parametric and Nonparametric ApproachesNow is probably a good time to compare and contrast the parametric approach to samplesize determination in a two-sample setting with the nonparametric approach. Notice werequire some similar pieces of information in both approaches: we require knowledge aboutthe level of the test, α, the desired effect, δ, and the number of observations per group, say,m. For the parametric comparison, we assume a common form of distribution for bothgroups, with an assumed known and identical scale parameter σ, and known locationparameters (means) μ1 and μ2, respectively. The nonparametric approach requires a smallpilot data set consisting of two groups without any assumptions about the commondistributional form. For pharmaceutical researchers, the nonparametric approach mayrequire more work because it requires data before the power may be calculated. However,the benefits of the procedure may greatly outweigh the expenditure of resources for thepilot.

First, investigators get to see the distribution of real data, as opposed to imagining whatit would look like, conditional on a value of location and scale and assuming somepre-specified mathematically convenient form. Often investigators will use the summarystatistics from a previously published manuscript to determine the power in the parametricapproach for their investigation. Were the originally published data distributed normally?Were they symmetrically distributed, for that matter? We really do not know by looking atthe summary statistics alone in the manuscript, nor can we learn much more if the authorsmade a poor choice of data visualization to summarize the paper’s findings. We only knowfor certain about the raw data’s features if we can see all of the data used in the originalmanuscript. It has been the author’s experience that such data are difficult to get, asscientific writers change institutional affiliations and are subsequently difficult to locateyears after they publish their research. Moreover, some are unwilling to share raw data, orunable to share it because of the proprietary nature of their research or because the dataare stored in an inconvenient form (e.g., in several spreadsheets or laboratory notebooks).

Second, the nonparametric approach is intuitive, given that a scientist understands howthe nonparametric test works. Power corresponds to the probability of, in some sense,finding a true and context meaningful difference, δ, at the α-level of significance for a givensample size. By employing the calculations described above, scientists can see where thevalue of power comes from without having to understand the mathematical statisticalproperties of the Gaussian (normal) distribution.

%KWSS MacroHow do we perform a nonparametric power analysis in SAS? The Mahoney-Magelgeneralization of the Collings-Hamilton approach to power determination in the k-samplesetting may be accomplished with a SAS macro called %KWSS provided on the book’scompanion Web site. The %KWSS macro consists of a body of code containing oneembedded macro (%BOOTSTRAP). The macro requires seven input parameters:

• IDSN is the name of an input SAS data set.• GROUP is the grouping or classification variable.• VAR is the response measurement variable.• SHIFTLIST is the shift vector, δ = (δ1, . . . , δk), for the test.

Page 161: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

148 Pharmaceutical Statistics Using SAS: A Practical Guide

• SAMPLE SIZE is the sample size for power determination.• ITERATION NO is the number of re-sampled data sets.• ALPHA is the desired level of statistical significance.

The macro first removes any observations that contain a missing response. It thendetermines the number of groups in the input data set IDSN and creates a list of thesegroups with the macro variable GRPVEC. The data are then subset by the various levelsof the classification variable. The embedded %BOOTSTRAP macro then performs therequired sampling (with replacement) of size SAMPLE SIZE*GROUP for each level ofGROUP. The location shifts specified in SHIFTLIST are applied to the re-sampled data byGROUP. If the number of location shifts specified in SHIFTLIST does not equal thenumber of groups or classes in the input data set, the program has a fail-safe that willterminate its execution. The macro then performs a Kruskal-Wallis test by PROCNPAR1WAY for each level of GROUP. The number of statistically significant (at levelALPHA) results is recorded as a fraction of the total number of re-sampled data sets(ITERATION NO) with PROC FREQ. For the GROUP levels of power determined,PROC MEANS determines the mean power and the result is reported via PROC PRINT.

To illustrate the usage of %KWSS, Program 6.15 estimates the power of theKruskal-Wallis test based on simulated data from a four-group experiment with 10observations/group. The response variable is assumed to follow a gamma distribution.Suppose that we are interested in using this pilot study information to power a study, alsowith four groups, but with a sample size allocation of 20 subjects/group. Suppose that weare interested in seeing a statistically significant (p < 0.05) shift in the location parameters,δ = (0, 1.5, 1.5, 1.5). The macro call is specified as follows:

%kwss(one,group,x,0 1.5 1.5 1.5,20,5000,0.05)

The first parameter is the name of the input data set (ONE) containing a responsevariable (X) and grouping or classification variable (GROUP) of interest (the second andthird macro parameters, respectively). The fourth parameter is the list of location shifts.The fifth parameter is the desired sample size/group. The sixth parameter is the number ofiterations (bootstrapped samples) per group of size 4 × 20 = 80. The final parameter is thedesired level of statistical significance for this four-sample test.

As a large number of bootstrap samples are requested for this power calculation,Program 6.15 will run for a considerable amount of time (about 20 minutes on the author’sPC using an interactive SAS session). The NONOTES system option is specified before theinvocation of the %BOOTSTRAP macro. Using this option prevents the annoyingsituation of a filled-up log in a PC SAS session that requires the user to empty or save thelog in a file.

Program 6.15 Nonparametric power calculation using the %KWSS macro

data one;do i=1 to 10;

x=5+rangam(1,0.5); group="A"; output;x=7+rangam(1,0.5); group="B"; output;x=7.2+rangam(1,0.5); group="C"; output;x=7.4+rangam(1,0.5); group="D"; output;drop i;

end;run;

%kwss(one,group,x,0 1.5 1.5 1.5,20,5000,0.05)

Page 162: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 6 Nonparametric Methods in Pharmaceutical Statistics 149

Output from Program 6.15

The Mahoney-Magel Generalization of the Collings-Hamilton Approach 1For Power Estimation with Location Shifts 0 1.5 1.5 1.5

of 4 Groups (A B C D) of Sample Size 20Compared at the 0.05-level of Statistical Significance

(Power Based on a Bootstrap Conducted 5000 Times)

POWER

99.945

Output 6.15 shows the estimated power of the Kruskal-Wallis test (99.945%). As wasthe case for other macros presented in this chapter, the titles of the output inform the userabout the information used to estimate the power (location shifts, number of groups,sample size, etc.).

6.4.4 SummaryThe approach to power estimation illustrated in this section differs from the classicalapproach where an investigator is required to provide estimates of treatment effect andvariation. In novel experimental investigations with limited resources, it may be difficult toprovide such estimates without executing a pilot study first. The approach, suggested firstby Collings and Hamilton (1988) and later generalized by Mahoney and Magel (1996), is areasonable approach to estimation of the power of an inference comparing locationparameters when estimates of treatment effect and variation are difficult to obtain. Theauthor has developed a macro to estimate power using the approach suggested by Mahoneyand Magel (the %KWSS macro).

AcknowledgmentsThe author is grateful to the following individuals for their contributions to this work: Dr.Dianne Camp (William Beaumont Research Institute) for her careful review and editorialsuggestions; Drs. Robert Abel and Cathie Spino (Pfizer Global Research and Development- Michigan Laboratories: Ann Arbor Campus) for their assistance with the clinical trialexamples; Ms Alice Birnbaum (Project Director, Axio Research) for her invaluablefeedback on the multiple comparison macros; and Dr. Thomas Vidmar (Senior Directorand Site Head, Midwest Nonclinical Statistics) for his support during this endeavor.

ReferencesAnsari, A.R., Bradley, R.A. (1960). “Rank-sum tests for dispersion.” Annals of Mathematical

Statistics. 31, 1174–1189.Bickel, P.J. Doksum, K.A. (1977). Mathematical Statistics: Basic Ideas and Selected Topics.

Oakland: Holden Day.Collings, B.J., Hamilton, M.A. (1988). “Estimating the power of the two-sample Wilcoxon test for

location shift.” Biometrics. 44, 847–860.Dunn, O.J. (1964). “Multiple comparisons using rank sums.” Technometrics. 6, 241–252.Dwass, M. (1960). “Some k-sample rank-order statistics.” Contributions to Probability and

Statistics. I. Olkin, S.G. Ghurye, H. Hoeffding, W.G. Madow, H.B. Mann, eds. Palo Alto:Stanford University Press, 198–202.

Fligner, M.A., Policello, G.E. (1981). “Robust rank procedures for the Behrens-Fisher problem.”Journal of the American Statistical Association. 76, 162–174.

Page 163: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

150 Pharmaceutical Statistics Using SAS: A Practical Guide

Harter, H.L. (1960). “Tables of range and studentized range.” Annals of Mathematical Statistics. 31,1122–1147.

Hochberg, Y., Tamhane, A.C. (1987). Multiple Comparison Procedures. New York: Wiley.Hollander, M., Wolfe, D.A. (1999). Nonparametric Statistical Methods. Second Edition. New York:

Wiley.Juneau, P. (2004). “Simultaneous nonparametric inference in a one-way layout using the SAS

System.” Proceedings of the PharmaSUG 2004 Annual Meeting. Available athttp://www.pharmasug.org/content/view/64/.

Kruskal, W.H., Wallis, W.A. (1952). “Use of ranks in one-criterion variance analysis.” Journal ofthe American Statistical Association. 47, 583–621.

Lehman, E.L. “Nonparametrics: Statistical Methods Based upon Ranks.” New York: Holden-Day,1975, pp. 206–207.

Mahoney, M., Magel, R. (1996). “Estimation of the power of the Kruskal-Wallis test.” BiometricalJournal. 38, 613–630).

Miller, R.G., Jr. (1966). Simultaneous Statistical Inference. First edition. New York: Springer-Verlag.Miller, R.G., Jr. (1981). Simultaneous Statistical Inference. Second edition. New York:

Springer-Verlag.National Asthma Council Australia. Asthma Management Handbook 2002. Available at

http://www.nationalasthma.org.au/html/management/amh/amh003.asp.Pillai, K.C.S., Ramachandran, K.V. (1954). “On the distribution of the ratio of the ith observation

in an ordered sample from a normal population to an independent estimate of the standarddeviation.” Annals of Mathematical Statistics. 25, 565–572.

Steel, R.G.D. (1960). “A rank sum test for comparing all pairs of treatments.” Technometrics. 2,197–207.

Student. (1908). “The probable error of a mean.” Biometrika. 6, 1–25.Wilcoxon, F. (1945). “Individual comparisons by ranking methods.” Biometrics. 1, 80–83.World Health Organization Fact Sheet No. 194 (revised January 2002). Available at

http://www.who.int/mediacentre/factsheets/fs194/en/.Zhou, X-H, Gao, S., Hui, S. L. (1997). “Methods for comparing the means of two independent

log-normal samples.” Biometrics. 53, 1129–1135.

Page 164: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7

Optimal Design ofExperiments inPharmaceutical Applications

Valerii FedorovRobert GagnonSergei LeonovYuehui Wu

7.1 Optimal Design Problem. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .152

7.2 Quantal Dose-Response Models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159

7.3 Nonlinear Regression Models with a Continuous Response . . . . . . . . . . . . . . . . . . . . . . . . . 165

7.4 Regression Models with Unknown Parameters in the Variance Function. . . . . . . . . . . . .169

7.5 Models with a Bounded Response (Beta Models) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172

7.6 Models with a Bounded Response (Logit Link) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176

7.7 Bivariate Probit Models for Correlated Binary Responses . . . . . . . . . . . . . . . . . . . . . . . . . . 181

7.8 Pharmacokinetic Models with Multiple Measurements per Patient . . . . . . . . . . . . . . . . . 184

7.9 Models with Cost Constraints . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190

7.10 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193

In this chapter we discuss optimal experimental designs for nonlinear models arising invarious pharmaceutical applications and present a short survey of optimal design methodsand numerical algorithms. We provide SAS code to implement optimal design algorithmsfor several examples:

• quantal models such as logistic models for analyzing success or failure in dose-responsestudies

• multi-parameter continuous logistic models in bioassays or pharmacodynamic studies,including models with unknown parameters in variance

• beta regression model

Valerii Fedorov is Group Director, Research Statistics Unit, Biomedical Data Sciences, GlaxoSmithKline Pharmaceuticals,USA. Robert Gagnon is Associate Director, Biomedical Data Sciences, GlaxoSmithKline Pharmaceuticals, USA. SergeiLeonov is Director, Research Statistics Unit, Biomedical Data Sciences, GlaxoSmithKline Pharmaceuticals, USA. YuehuiWu is Senior Statistician, Research Statistics Unit, Biomedical Data Sciences, GlaxoSmithKline Pharmaceuticals, USA.

Page 165: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

152 Pharmaceutical Statistics Using SAS: A Practical Guide

• models with multiple responses, such as models that measure both efficacy and safety indose response studies and pharmacokinetic models with multiple samples per subject

• models with cost constraints.

For all examples, we use a first-order optimization algorithm in the space of informationmatrices. A short survey of other software tools for constructing optimal model-baseddesigns is provided.

7.1 Optimal Design ProblemWe start this chapter with the description of the general optimal design problems andconcepts.

7.1.1 General ModelConsider a vector of observations Y = {y1, . . . , yN} and assume it follows a generalparametric model. The joint probability density function of Y depends on x and θ, where xis the independent, or design, variable and θ = (θ1, . . . , θm) is the vector of unknown modelparameters. The design variable x is chosen, or controlled, by researchers to obtain the bestestimates of the unknown parameters.

This general model can be applied to a wide variety of problems arising in clinical andpre-clinical studies. The examples considered in this chapter are described below.

Dose-Response StudiesDose-response models arise in clinical trials, either with a binary outcome (e.g.,success-failure or dead-alive in toxicology studies; see Section 7.2) or continuous response(e.g., studies of pain medications when patients mark their pain level on a visual analogscale; see Section 7.4). In these examples, x represents the dose of a drug administered tothe patient. Figure 7.1 illustrates the dependence of the probability of the success π(x) atthe dose x for a two-parameter logistic model. Dotted horizontal lines correspond to theprobabilities of success for the two optimal doses x∗

1 and x∗2. See Section 7.2 for details.

Figure 7.1 Optimal design points (vertical lines) in a two-parameter logistic model

Prob=0.824

Prob=0.176

Pro

babi

lity

of r

espo

nse

0.0

0.5

1.0

Dose on a log scale

-2.0 -1.5 -1.0 -0.5 0.0 0.5 1.0

Page 166: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 153

Bioassay StudiesMulti-parameter logistic models, sometimes referred to as the Emax or Hill models, arewidely used in bioassay studies. Examples include models that relate the concentration ofan experimental drug to the percentage or number of surviving cells in cell-based assays ormodels that quantify the concentration of antigens or antibodies in enzyme-linkedimmunosorbent assays (ELISA). In this context, the design variable x represents the drugconcentration level; see Sections 7.2 and 7.3 for details.

As an illustration, Figure 7.2 plots a four-parameter logistic model that describes thenumber of surviving bad cells in a cell-based assay versus drug concentration x on thelog-scale. The concentration-effect relationship is negative. This is expected since anincrease in the drug concentration usually reduces the number of cells.

Figure 7.2 Four-parameter logistic model

Cel

l cou

nt

400

800

1200

1600

Concentration on a log scale

0 1 2 3 4 5 6

Two-Drug Combination StudiesSection 7.6.3 discusses two-drug combination studies in which drugs (e.g., Drug A andDrug B) are administered simultaneously and their effect on the patient’s outcome isanalyzed. In this case, the design variable x is two-dimensional, i.e., x = (x1, x2), where x1is the dose of Drug A and x2 is the dose of Drug B.

Clinical Pharmacokinetic StudiesMultiple blood samples are taken in virtually all clinical pharmacokinetic (PK) studies andthe collected data are analyzed by means of various PK compartmental models. Thisanalysis leads to quite sophisticated nonlinear mixed effects models which are discussed inSection 7.8. In these models x is a k-dimensional vector that represents a collection(sequence) of sampling times for a particular patient.

Cost-Based DesignsIn the previous example (PK studies) it is quite obvious that each extra sample providesadditional information. On the other hand, the number of samples that may be drawn fromeach patient is restricted because of blood volume limitations and other logistic and ethicalreasons. Moreover, the analysis of each sample is associated with monetary cost. Thus, it

Page 167: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

154 Pharmaceutical Statistics Using SAS: A Practical Guide

makes sense to incorporate costs in the design. One potential approach for the constructionof cost-based designs is discussed in Section 7.9.

SAS Code and Data Sets

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

7.1.2 Information Matrix, Maximum Likelihood Estimates, andOptimality Criteria

For any parametric model, one of the main goals is to obtain the most precise estimates ofunknown parameters θ. For example, in the context of a dose-response study, if clinicalresearchers can select doses xi, in some admissible range, and number of patients ni oneach dose, then, given the total number of observations N , the question is how to allocatethose doses and patients to obtain the best estimates of unknown parameters. The qualityof estimators is traditionally measured by their variance-covariance matrix.

To introduce the theory underlying optimal design of experiments, we will assume thatni is the number of independent observations yij that are made at design point xi and thatN = n1 + n2 + . . . + nn is the total number of observations, j = 1, . . . , ni, i = 1, . . . , n. Letthe probability density function of yij be p(yij |xi, θ). For example, in the context of adose-response study, xi is the ith dose of the drug and yij is the response of the jth patienton dose xi.

Most of the examples discussed in this chapter deal with the case of a singlemeasurement per patient, i.e., both xi and yij are scalars (single dose and single response)and yij and yi1,j1 are independent if i �= i1 or j �= j1. The exceptions are in Section 7.7 (twobinary responses for the same patient, such as efficacy and toxicity, at a given dose xi) andSections 7.8 and 7.9 that consider the problem of designing experiments with multiple PKsamples over time for the same patient, i.e., a design point is a sequencexi = (x1

i , x2i , . . . , x

ki ) of k sampling times and yij = (y1

ij , . . . , ykij) is a vector of k observations.

Note that in the latter case the elements of vector yij are, in general, correlated becausethey correspond to measurements on the same patient. However, similar to the case of asingle measurement per patient, yij and yi1,j1 are still independent if i �= i1 or j �= j1.

Information Matrix

Let μ(x, θ) be an m × m information matrix of a single observation at point x,

μ(x, θ) = E

[∂ log p(Y |x, θ)

∂θ

∂ log p(Y |x, θ)∂θT

], (7.1)

where the expectation is taken with respect to the distribution of Y . The Fisherinformation matrix of the N experiments can be written as

MN(θ) =n∑

i=1

niμ(xi, θ). (7.2)

Further, let M(ξ, θ) be a normalized information matrix,

M(ξ, θ) =1N

MN(θ) =n∑

i=1

wiμ(xi, θ), wi =ni

N. (7.3)

The collection ξ = {xi, wi} is called a normalized (continuous or approximate) designwith w1 + . . . + wn = 1. In this setting N may be viewed as a resource available toresearchers; see Section 7.9 for a different normalization.

Page 168: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 155

Maximum Likelihood EstimatesThe maximum likelihood estimate (MLE) of θ is given by

θ = arg minθ

n∏i=1

ni∏j=1

p(yij |xi, θ) = arg minθ

n∑i=1

ni∑j=1

log p(yij |xi, θ),

It is well known that the variance-covariance matrix of θ for large samples is approximatedby

Var(θ) ≈ M−1N (θ) =

1N

M−1(ξ, θ) =1N

D(ξ, θ), (7.4)

where D(ξ, θ) = M−1(ξ, θ) is the normalized variance-covariance matrix (Rao, 1973,Chapter 5). Throughout this chapter we assume that the normalized information matrixM(ξ, θ) is not singular and thus its inverse exists.

Optimality CriteriaIn the convex design theory, it is standard to minimize various functionals depending onD(ξ, θ),

ξ∗ = arg minξ

Ψ[D(ξ, θ)], (7.5)

where Ψ is a selected functional (criterion of optimality). The optimization is performedwith respect to designs ξ,

ξ = {wi, 0 ≤ wi ≤ 1,n∑

i=1

wi = 1; xi ∈ X},

where X is a design region, e.g., admissible doses in a dose-response study, and the weights,w1, . . . , wn, are continuous variables. The use of continuous weights leads to the concept ofapproximate optimal designs; the word approximate should draw attention to the fact thatthe solution of (7.5) does not generally give integer values ni = Nwi. However, this solutionis often acceptable, in particular when the total number of observations N is relativelylarge and we round ni = Nwi to the nearest integer while keeping n1 + . . . + nn = N(Pukelsheim, 1993).

The following optimality criteria are among the most popular ones:

D-optimality. Ψ = ln |D(ξ, θ)|, where |D| denotes the determinant of D. This criterion isoften called a generalized variance criterion since the volume of the confidence ellipsoidfor θ is proportional to |D(ξ, θ)|1/2; see Fedorov and Hackl (1997, Chapter 2).

A-optimality. Ψ = tr[AD(ξ, θ)], where A is an m × m non-negative definite matrix (utilitymatrix) and tr(D) denotes the trace, or sum of diagonal elements, of D. For example, ifA = m−1Im, where Im is an m × m identity matrix, the A-criterion is based on theaverage variance of the parameter estimates:

Ψ = m−1m∑

i=1

Var(θi).

c-optimality. Ψ = cT D(ξ, θ)c, where c is an m-dimensional vector. A c-optimal designminimizes the variance of a linear combination of the model parameters, i.e., cT θ.

E -optimality. Ψ = λmin[M(ξ, θ)] = λmax[D(ξ, θ)], where λmin(M) and λmax(M) are minimaland maximal eigenvalues of M , respectively. Note that the length of the largest principalaxis of the confidence ellipsoid is λ

−1/2min [M(ξ, θ)].

Page 169: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

156 Pharmaceutical Statistics Using SAS: A Practical Guide

I -optimality. Ψ =∫

X tr[μ(x, θ)D(ξ, θ)]dx. In linear regression models, tr[μ(x, θ)D(ξ, θ)]represents the variance of the predicted response.

Minimax optimality. Ψ = maxx∈X tr[μ(x, θ)D(ξ, θ)].

For other criteria of optimality, see Fedorov (1972), Silvey (1980), or Pukelsheim (1993).It is important to note that the D-, A-, and E-optimality criteria may be ordered for

any design since

|D|1/m ≤ m−1tr(D) ≤ λmax(D), D = D(ξ, θ),

see Fedorov and Hackl (1997, Chapter 2). Also, the disadvantage of c-optimal designs isthat they are often singular, i.e., the corresponding variance-covariance matrix isdegenerate.

D-Optimality Criterion

In this chapter we concentrate on the D-optimality criterion (note that the SAS programsdescribed in this chapter can also be used to construct A-optimal designs). D-optimaldesigns are popular among theoretical and applied researchers due to the followingconsiderations:

• D-optimal designs minimize the volume of the asymptotic confidence region for θ. Thisproperty is easy to explain to practitioners in various fields.

• D-optimal designs are invariant with respect to non-degenerate transformations ofparameters (e.g., changes of the parameter scale).

• D-optimal designs are attractive in practice because they often perform well accordingto other optimality criteria; see Atkinson and Donev (1992, Chapter 11) or Fedorov andHackl (1997).

7.1.3 Locally Optimal DesignsIt is easy to see from the definition of the information matrix M(ξ, θ) that, in general,optimal designs depend on the unknown parameter vector θ. This assumption leads to theconcept of locally optimal designs that are defined as follows: first we need to specify apreliminary estimate of θ, e.g., θ, and then solve the optimization problem for the given θ(Chernoff, 1953, Fedorov, 1972).

It is worth noting that in linear parametric models the information matrix does notdepend on θ, which greatly simplifies the problem of computing optimal designs. Consider,for example, a linear regression model

yij = θ1f1(xi) + . . . + θmfm(xi) + εij , i = 1, . . . , n, j = 1, . . . , ni

with normally distributed residuals, i.e., εij ∼ N(0, σ2(xi)). Here f(x) = [f1(x), . . . , fm(x)]Tis a vector of pre-defined basis functions. The information matrix is given by (Fedorov andHackl, 1997)

μ(x) = σ−2(xi)f(x)fT (x)

and thus we can construct designs that will be optimal for any value of θ. In non-linearmodels, we need to select a value of θ first; however, once this value is fixed, theconstruction of an optimal design is absolutely the same as for linear problems.

Obviously, the quality of locally optimal designs may be poor if the preliminary estimateθ significantly differs from the true value of θ. We mention in passing that this problem canbe tackled by using various techniques which include minimax designs (Fedorov and Hackl,

Page 170: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 157

1997), Bayesian designs (Chaloner and Verdinelli, 1995), adaptive designs (Box andHunter, 1965; Fedorov, 1972, Chapter 4; Zacks, 1996, Fedorov and Leonov, 2005, Section5.6). While minimax and Bayesian approaches take into account prior uncertainties, theylead to optimization problems which are computationally more demanding than theconstruction of locally optimal designs. Instead of preliminary estimates of unknownparameters, we must provide an uncertainty set for the minimax approach and a priordistribution for Bayesian designs. The latter task is often based on a subjective judgment.Locally optimal designs serve as a reference point for other candidate designs, andsensitivity analysis with respect to parameter values is always required to validate theproperties of a particular optimal design; see a discussion in Section 7.3.1.

In this chapter we concentrate on the construction of locally optimal designs.

7.1.4 Equivalence TheoremMany theoretical results and numerical algorithms of the optimal experimental designtheory rely on the following important theorem (Kiefer and Wolfowitz, 1960; Fedorov,1972, White, 1973):

Generalized equivalence theorem. A design ξ∗ is locally D-optimal if and only if

ψ(x, ξ∗, θ) = tr[μ(x, θ)M−1(ξ∗, θ)

]≤ m, (7.6)

where m is the number of model parameters. Similarly, a design ξ∗ is locally A-optimal ifand only if

ψ(x, ξ∗, θ) = tr[μ(x, θ)M−1(ξ∗, θ)AM−1(ξ∗, θ)

]≤ tr

[AM−1(ξ∗, θ)

]. (7.7)

The equality in (7.6) and (7.7) is attained at the support points of the optimal design ξ∗.The ψ(x, ξ, θ) function is termed the sensitivity function of the corresponding criterion.

The sensitivity function helps identify design points that provide the most informationwith respect to the chosen optimality criterion (Fedorov and Hackl, 1997, Section 2.4). Forinstance, if we consider dose-response studies, optimal designs can be constructediteratively by choosing doses x∗ that maximize the sensitivity function ψ(x, ξ, θ) over theadmissible range of doses.

As shown in the next subsection, the general formulas (7.6) and (7.7) form a basis ofnumerical procedures for constructing D- and A-optimal designs.

7.1.5 Computation of Optimal DesignsAny numerical procedure for constructing optimal designs requires two key elements:

• The information matrix μ(x, θ) or, equivalently, sensitivity function ψ(x, ξ, θ).• The design region X (the set of admissible design points).

In all examples discussed in this chapter (except for the examples considered in Sections7.8 and 7.9), we define the design region as a compact set, but search for optimal points ona pre-defined discrete grid. This grid can be rather fine in order to guarantee that theresulting design is close to the optimal one.

The main idea behind the general nonlinear design algorithm is that, at each step of thealgorithm, the sensitivity function ψ(x, ξ, θ) is maximized over the design region todetermine the best new support point (forward step) and then minimized over the supportpoints of the current design to remove the worst point in the current design (backwardstep). See Fedorov and Hackl (1997) or Atkinson and Donev (1992) for details.

Page 171: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

158 Pharmaceutical Statistics Using SAS: A Practical Guide

To define a design algorithm, let ξs = {Xis, wis}, i = 1, . . . , ns, be the design at Step s.Here {Xis} is the vector of support points in the current design and wis is the vector ofweights assigned to Xis. The iterative algorithm is of the following form:

ξs+1 = (1 − αs)ξs + αsξ(Xs),

where ξ(X) is a one-point design supported on point X.

Forward step. At Step s, a point X+s that maximizes ψ(x, ξ, θ) over x ∈ X is added to the

design ξs with weight αs = γs, where γs = 1/(n0 + s) and n0 is the number of points inthe initial design.

Backward step. After that, a point X−s that minimizes ψ(x, ξ, θ) over all support points

in the current design is deleted from the design with weight

αs ={

−γs, ws ≥ γs,−ws/(1 − ws), ws < γs.

In general, the user can change γs to c1/(n0 + c2s), where c1 and c2 are two constants.The default values of the constants are c1 = c2 = 1.

In Section 7.2 we provide a detailed description of a SAS macro that implements thedescribed optimal design algorithm for quantal dose-response models. However, once theinformation matrix μ(x, θ) or the sensitivity function ψ(x, ξ, θ) is specified, the sametechnique can be used to generate optimal designs for any other model.

7.1.6 Existing SoftwareComputer algorithms for generating optimal designs have existed for quite some time(Fedorov, 1972; Wynn, 1970). These algorithms have been implemented in manycommercially available software systems. Additionally, software written by academic groupsis available. In general, the commercially available systems implement methods forproblems such as simple linear regression and factorial designs, but not for more complexmodels such as nonlinear regression, beta regression, or population pharmacokinetics.Academic groups have developed and made available programs for the latter cases.

On the commercial side, SAS offers both SAS/QC (the OPTEX procedure) and JMP.The OPTEX procedure supports A- and D-optimal designs for simple regression models.JMP generates D-optimal factorial designs, as do software packages such as Statistica. Inpharmacokinetic applications, the ADAPT II program developed at the University ofSouthern California implements c- and D-optimal and partially optimal design generationfor individual pharmacokinetic models; however, it does not support more complexpopulation pharmacokinetic models (D’Argenio and Schumitzky, 1997). Ogungbenro et al.(2005) implemented in Matlab the classical exchange algorithm for population PKexperiments using D-optimality (note that the exchange algorithm improves the initialdesign with respect to selected optimality criterion but, in general, does not converge tothe optimal design). Retout and Mentre (2003) developed extensive implementation ofpopulation pharmacokinetic designs in S-Plus.

In this chapter we discuss SAS/IML implementation of the general algorithm (whichcannot be executed in PROC OPTEX) with applications to several widely used nonlinearmodels. We also describe optimal design algorithms for population pharmacokinetic modelsanalogous to the S-Plus programs of Retout and Mentre (2003).

Page 172: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 159

7.1.7 Structure of SAS ProgramsThere are five components in the programs:

• inputs• establishment of the design region• calculation of the information matrix• using the optimal design algorithm• outputs.

Only the optimal design algorithm component is unchanged for any model; the othercomponents are model-specific and need to be modified accordingly. In any model,calculation of the information matrix is the critical (and most computationally intensive)step.

7.2 Quantal Dose-Response ModelsQuantal, or binary, models arise in various pharmaceutical applications, such as clinicaltrials with a binary outcome, toxicology studies, and quantal bioassays.

7.2.1 General ModelIn a quantal model, a binary response variable Y depends on the dose x,

Y = Y (x) ={

1, a response is present at dose x,0, no response,

and the probability of observing a response is modeled as

P{Y = 1|x} = η(x, θ),

where 0 ≤ η(x, θ) ≤ 1 is a given function and θ is a vector of m unknown parameters. It isoften assumed that

η(x, θ) = π(z),

where z is a linear combination of pre-defined functions of the dose x, i.e.,

z = θ1f1(x) + . . . + θmfm(x).

In many applications, π(z) is selected as a probability distribution function with acontinuous derivative (see Fedorov and Leonov, 2001). Among the most popular choices ofthis function are:

Logistic (logit) model. The π(z) function is a logistic function

π(z) = ez/(1 + ez).

Probit model. π(z) is a standard normal cumulative distribution function,

π(z) =1√2π

∫ z

−∞e−u2/2du.

In practice, when properly normalized, the two models lead to virtually identical results(Cramer, 1991, Section 2.3; Finney, 1971, p. 98).

Page 173: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

160 Pharmaceutical Statistics Using SAS: A Practical Guide

In the quantal dose-response model the probability density function of Y at a point x isgiven by

p(Y |x, θ) = η(x, θ)Y [1 − η(x, θ)]1−Y

and thus the information matrix of a single observation is equal to

μ(x, θ) =[π′(z)]2

π(z)[1 − π(z)]f(x)fT (x),

(Wu, 1988, Torsney and Musrati, 1993) where f(x) = [f1(x), . . . , fm(x)]T and thus thevariance-covariance matrix of the MLE, θ, can be approximated by D(ξ, θ)/N ; see (7.4).

The quantal logistic model provides an example where the statement of the generalizedequivalence theorem, (7.6) and (7.7), admits a more “numerically-friendly” presentation.For example, the information matrix for a single observation can be written asμ(x, θ) = g(x, θ)gT (x, θ), where

g(x, θ) =π′(z)f(x)√

π(z)[1 − π(z)],

which, together with the matrix identity tr(AB) = tr(BA), leads to the followingpresentation of the sensitivity functions for the quantal model:

D-criterion: ψ(x, ξ, θ) = g(x, θ)M−1(ξ, θ)gT (x, θ), (7.8)

A-criterion: ψ(x, ξ, θ) = g(x, θ)M−1(ξ, θ)AM−1(ξ, θ)gT (x, θ). (7.9)

7.2.2 Example 1: A Two-Parameter Logistic ModelIn this subsection we will take a closer look at the logistic model with two unknownparameters:

P{Y = 1|x} = ez/(1 + ez), z = θ1 + θ2x,

where θ1 and θ2 are the intercept and slope parameters, respectively.It can be shown that D-optimal designs associated with the described model are

two-point designs, with half the measurements at each dose, i.e., w1 = w2 = 0.5 (White,1975). It is interesting to note that, when the dose range is sufficiently wide, D-optimaldesigns are uniquely defined in the z-space and the optimal design points correspond tocertain response probabilities. Specifically, the optimal design points on the z scale arezopt = ±1.543 and the corresponding response probabilities are given by

π(−1.543) = 0.176, π(1.543) = 0.824,

where π(z) = ez/(1 + ez). Thus, if x∗1 and x∗

2 are the two optimal doses corresponding to aD-optimal design, then

θ1 + θ2x∗1 = −1.543, θ1 + θ2x

∗2 = 1.543.

It is also worth pointing out that, if xp is a dose level that causes a particular responsein 100p% of subjects, i.e., η(xp, θ) = p, the normalized variance of the MLE xp in thetwo-parameter logistic model is a special case of the c-optimality criterion and can bewritten as

Ψ = Var(xp) = cTp D(ξ, θ)cp = tr

[(cpc

Tp

)D(ξ, θ)

](Wu, 1988) with cp = f(xp)/θ2. For a discussion of c- and A-optimal designs for binarymodels, see Ford, Torsney and Wu (1992), Sitter and Wu (1993).

Page 174: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 161

IllustrationAssume that the true values of the two parameters in the logistic model are θ1 = 1 andθ2 = 3. Assume also that the admissible dose range (design region) is given byX = [xmin, xmax] with xmin = −1 and xmax = 1. First of all, note that

1 + 3xmin < −1.543, 1 + 3xmax > 1.543

and therefore the D-optimal dose levels lie inside the design region. The optimal doses,(x∗

1, x∗2), are found from

1 + 3x∗1 = −1.543, 1 + 3x∗

2 = 1.543.

It is easy to verify that x∗1 = −0.848 and x∗

2 = 0.181. The associated weights arew1 = w2 = 0.5.

Computation of the D-Optimal Design

This subsection introduces the %OptimalDesign1 macro that implements the optimaldesign algorithm in the univariate case (a single design variable x) and illustrates theprocess of computing a D-optimal design for the two-parameter logistic model describedabove. The advantage of using this simple model is that we can easily check individualelements of the resulting optimal design, including the information matrix, optimal doses,and weights.

The %OptimalDesign1 macro supports two main components of the optimal designalgorithm:

• calculation of the information matrix (%deriv and %infod macros)• implementation of the forward and backward steps of the algorithm (%doptimal macro)

The first component (information matrix) in this list is model-specific, whereas thesecond component (algorithm) does not depend on the chosen model. Note that the firstcomponent is the critical (and most computationally intensive) step in this macro.

Program 7.1 invokes the %OptimalDesign1 macro to compute a D-optimal design forthe two-parameter logistic model. To save space, the complete SAS code is provided on thebook’s companion Web site. This subsection focuses on the user-specified parameters thatdefine the design problem and set up the optimal design algorithm.

The first four macro variables in Program 7.1 define the following design parameters:

• POINTS is a vector of doses included in the initial design. In this case, the initial designcontains four dose levels evenly spaced across the dose range X = [−1, 1].

• WEIGHTS defines the weights of the four doses in the initial design (the doses areequally weighted).

• GRID defines the grid points in the optimal design algorithm. The grid consists of 201equally spaced points in the dose range. The DO function in SAS/IML creates a list ofequally spaced points. In this case, DO produces the following row vector,{−1,−0.99,−0.98, . . . , 0.98, 0.99, 1}.

• PARAMETER is a vector of model parameters. The true values of θ1 and θ2 areassumed to be 1 and 3, respectively.

The other five macro variables define the algorithm properties of the optimal designalgorithm:

• CONVC is the convergence criterion. It determines when the iterative procedureterminates.

Page 175: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

162 Pharmaceutical Statistics Using SAS: A Practical Guide

• MAXIMIT is the maximum number of iterations. A typical value of MAXIMIT is 200.A greater value is recommended when the final design is expected to contain a largenumber of points.

• CONST1 and CONST2 are used in the calculation of weights in the optimal designalgorithm (Section 7.1.5). By default, these constants are set to 1. The user can changethese values to facilitate the convergence of the algorithm in models with a large numberof parameters.

• CMERGE is the merging constant in the optimal design algorithm that influences theprocess of merging design points. The default value of CMERGE is 3 and a larger valueshould be considered if the final design includes points with very small weights.

To calculate the information matrix μ(x, θ) and the sensitivity function ψ(x, ξ, θ), weneed to specify the g(x, θ) function introduced in Section 7.2.1. This function is defined inthe %deriv macro. The MATRIX variable is the vector that contains the values of x, andthe vector representing the g(x, θ) function is stored in the DERIVATIVE variable. In thiscase, the g(x, θ) function is given by

g(x, θ) =ez/2

1 + ezf(x), f(x) = (1, x)T .

Program 7.1 D-optimal design for the two-parameter logistic model (Design parameters, algorithmparameters and g(x, θ) function)

* Design parameters;%let points={-1 -0.333 0.333 1};%let weights={0.25 0.25 0.25 0.25};%let grid=do(-1,1,0.01);%let parameter={1 3};* Algorithm parameters;%let convc=1e-7;%let maximit=200;%let const1=1;%let const2=1;%let cmerge=3;* G function;%macro deriv(matrix,derivative);

nm=nrow(&matrix);one=j(nm,1,1);fm=one||&matrix;gc=exp(0.5*fm*t(parameter))/(1+exp(fm*t(parameter)));&derivative=gc#fm;

%mend deriv;* Optimal design algorithm;%OptimalDesign1;

Output from Program 7.1

Initial design

Weight X

0.250 -1.0000.250 -0.3330.250 0.3330.250 1.000

Page 176: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 163

Optimal design

Weight X

0.501 -0.8500.499 0.180

Determinants of the variance-covariance matrices

INITIAL OPTIMAL

271.1 179.6

Variance-covariance matrix, initial design

COL1 COL2

11.0 9.29.2 32.4

Variance-covariance matrix, optimal design

COL1 COL2

9.8 8.78.7 26.0

The output produced by Program 7.1 includes the following data sets (Output 7.1):

• INITIAL data set (design points and associated weights in the initial design).• OPTIMAL data set (design points and associated weights in the D-optimal design).• DDET data set (determinants of the variance-covariance matrix D(ξ, θ) for the initial

and optimal designs).• DINITIAL data set (variance-covariance matrix D(ξ, θ) for the initial design).• DOPTIMAL data set (variance-covariance matrix D(ξ, θ) for the optimal design).

The program also creates two plots (Figure 7.3):

• Plot of the initial and optimal designs.• Plot of the sensitivity functions associated with the initial and optimal designs.

Output 7.1 shows that the optimal doses are x∗1 = −0.85 and x∗

2 = 0.18 with equalweights (see also the left panel in Figure 7.3). This example illustrates a well-knowntheoretical result that if a D-optimal design is supported at m points for a model with munknown parameters, then the support points have equal weights, wi = 1/m, i = 1, . . . , m;see Fedorov (1972, Corollary to Theorem 2.3.1). It is easy to check that the optimal dosesare very close to the doses we computed earlier from the equation zopt = ±1.543.

We can also see from Output 7.1 that the determinant of the optimalvariance-covariance matrix is 179.6 compared to the initial value of 271.1. There was alsoan improvement in the variance of the parameter estimates. The variance of θ1 droppedfrom 11.0 to 9.8 and the variance of θ2 decreased from 32.4 to 26.0.

The right panel in Figure 7.3 shows that the equivalence theorem serves as an excellentdiagnostic tool in optimization problems. The sensitivity function of the D-optimal designhits the reference line m = 2 at the optimal doses, i.e., at x = −0.85 and x = 0.18. For the

Page 177: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

164 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 7.3 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivity func-tions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Dose

-1.0 -0.5 0.0 0.5 1.0

Sen

sitiv

ity

0

1

2

3

Dose

-1.0 -0.5 0.0 0.5 1.0

D-criterion, the sensitivity function is identical to the normalized variance of prediction.Thus, since the sensitivity function of the initial design is greater than m = 2 at the leftend of the dose range, we conclude that

• The initial design is not D-optimal.• More measurements should be placed at the left end of the design region in order to

reduce the variance of prediction or, equivalently, push the sensitivity function downbelow the reference line defined by the equivalence theorem (m = 2).

7.2.3 Example 2: A Two-Parameter Logistic Model with a Narrow Dose RangeIn Example 1, we considered the case when the admissible dose range was fairly wide,X = [−1, 1]. If the dose range is not sufficiently wide, at least one of the D-optimal pointsmay coincide with the boundary and we will no longer be able to use the simple ruleintroduced earlier in this section (zopt = ±1.543).

As an illustration, consider a narrower dose range, X = [0, 1]. To compute the D-optimaldesign for the two-parameter logistic model in this case, all we need to do is to modify thelower boundary of the dose range in Program 7.1. Program 7.2 derives the D-optimaldesign for the modified dose range (complete SAS code is given on the book’s companionWeb site).

Program 7.2 D-optimal design for the two-parameter logistic model with a narrow dose range (Algorithmparameters and g(x, θ) function are identical to those defined in Program 7.1)

* Design parameters;%let points={0 0.333 0.667 1};%let weights={0.25 0.25 0.25 0.25};%let grid=do(0,1,0.025);%let parameter={1 3};

Page 178: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 165

Output 7.2

Initial design

Weight X

0.250 0.0000.250 0.3330.250 0.6670.250 1.000

Optimal design

Weight X

0.500 0.0000.500 0.725

Output 7.2 shows that the resulting D-optimal design is still a two-point design withequal weights w1 = w2 = 0.5; however, the optimal doses have changed. One dose is nowlocated at the boundary of the region, x∗

1 = 0, and the other dose has shifted to the right,x∗

2 = 0.725 (see also Figure 7.4). As was pointed out in Section 7.1.5, optimal designsdepend not only on the information matrix (which is identical to the information matrix inExample 1) but also on the dose range or, in general, the design region.

Figure 7.4 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivity func-tions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Dose

0.00 0.25 0.50 0.75 1.00

Sen

sitiv

ity

0

1

2

3

4

Dose

0.00 0.25 0.50 0.75 1.00

7.3 Nonlinear Regression Models with a Continuous ResponseIn this section we first formulate the optimal design problem for general nonlinearregression models and then consider a popular dose-response model with a continuousresponse variable (four-parameter logistic, or Emax, model).

7.3.1 General Nonlinear Regression ModelConsider a general nonlinear regression model that is used in the analysis of concentration-or dose-response curves in a large number of clinical and pre-clinical studies. The model

Page 179: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

166 Pharmaceutical Statistics Using SAS: A Practical Guide

describes the relationship between a continuous response variable, Y , and design variable x(dose or concentration level):

E(yij |xi; θ) = η(xi, θ), i = 1, . . . , n, j = 1, . . . , ni.

Here yij ’s are often assumed to be independent observations with Var(yij |xi; θ) = σ2(xi),i.e., the variance of the response variable varies across doses or concentration levels. Also,xi is the design variable that assumes values in a certain region, X . Lastly, θ is anm-dimensional vector of unknown parameters.

When the response variable Y is normally distributed, the variance matrix of the MLE,θ, can be approximated by D(ξ, θ)/N , see (7.4), and the information matrix of a singleobservation is

μ(xi, θ) = g(xi, θ)gT (xi, θ), g(xi, θ) =f(xi, θ)σ(xi)

,

where f(x, θ) is a vector of partial derivatives of the response function η(x, θ),

f(x, θ) =[∂η(x, θ)

∂θ1,∂η(x, θ)

∂θ2, . . . ,

∂η(x, θ)∂θm

]T

. (7.10)

Now, as far as optimal experimental design is concerned, the sequence of steps that followis exactly the same as for quantal dose-response models that are considered in Section 7.2:

1. Select a preliminary parameter estimate, θ.2. Select an optimality criterion, Ψ = Ψ[D(ξ, θ)], e.g., the D-optimality criterion.

3. Construct a locally optimal design ξ∗(θ) for the selected criterion.

It is recommended that sensitivity analysis be performed, i.e., steps 1 through 3 must berepeated for slightly different θs, s = 1, . . . , Ns, to verify robustness of the design ξ∗(θ); seeAtkinson and Fedorov (1988).

Since we know that μ(xi, θ) = g(xi, θ)gT (xi, θ), the process of constructing D- orA-optimal designs for the general nonlinear regression model will be virtually identical tothe process of finding optimal designs for quantal dose-response models (described in detailin Section 7.2). We need only to redefine the g(x, θ) function that determines theinformation matrix μ(x, θ) and sensitivity functions in (7.6) and (7.7) for the D- andA-optimality criteria, respectively.

7.3.2 Example 3: A Four-Parameter Logistic Model with aContinuous Response

The four-parameter logistic, or Emax, model, is used in many pharmaceutical applications,including bioassays (of which ELISA, or enzyme-linked immunosorbent assay, andcell-based assays are popular examples). See Finney (1976), Karpinski (1990), Hedayatet al. (1997), Kallen and Larsson (1999) for details and references.

This example comes from a study of a compound that inhibits the proliferation of bonemarrow erythroleukemia cells in a cell-based assay (Downing, Fedorov and Leonov, 2001;Fedorov and Leonov, 2005). The logistic model used in the study is defined as follows:

E(Y |x, θ) = η(x, θ) = θ3 +θ4 − θ3

1 + (x/θ1)θ2,

where Y is a continuous outcome variable (number of cells in the assay) and x is the designvariable (concentration level). Further, θ1 is often denoted as ED50 and represents the

Page 180: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 167

concentration at which the median response (θ3 + θ4)/2 is attained, θ2 is a slope parameter,θ4 and θ3 are the lower and upper asymptotes (minimal and maximal responses).

As an aside note, the four-parameter logistic model provides an example of a partiallynonlinear model since θ3 and θ4 enter the response function in a linear fashion. This linearentry implies that the D-optimal design does not depend on values of θ3 and θ4 (Hill, 1980).

Computation of the D-Optimal DesignThe vector of unknown parameters θ was estimated by modeling the data collected from a96-well plate assay, with six repeated measurements at each of the ten concentrations,{500, 250, 125, . . . , 500/29} ng/ml, which represents a two-fold serial dilution design withN = 60 and weights wi = 1/10. The estimated parameters are

θ = (15.03, 1.31, 530, 1587)T .

The design region will be set to

X = [ln(500/29), ln(500)] = [−0.024, 6.215]

on a log-scale. The x values will be exponentiated to compute the response function η(x, θ).Program 7.3 constructs the D-optimal design for the introduced four-parameter logistic

model by calling the %OptimalDesign1 macro. As in Programs 7.1 and 7.2, we need todefine the following parameters and pass them to the macro:

• The initial design is based on ten equally spaced and equally weighted log-transformedconcentration levels within the design region (POINTS and WEIGHTS variables).

• The grid consists of 801 equally spaced points in X = [−0.024, 6.215].• The true values of the model parameters are specified using the PARAMETER variable.• The algorithm parameters are identical to those used in Programs 7.1 and 7.2.• To specify the g(x, θ) function, we need to compute a vector of partial derivatives. The

derivatives are defined in the %deriv macro. The MATRIX variable contains the valuesof x. The DER1, DER2, DER3, and DER4 variables represent the derivatives of theresponse function with respect to θ1, θ2, θ3, and θ4, respectively, and the DERIVATIVEvariable contains the vector of partial derivatives.

Also, note that in order to improve the stability of the iterative procedure, the mergingconstant (CMERGE) needs to be increased to 12.

As before, we will concentrate on the user-defined parameters. The complete SAS codecan be found on the book’s companion Web site.

Program 7.3 D-optimal design for the four-parameter logistic model with a continuous response (Designparameters, algorithm parameters and g(x, θ) function)

* Design parameters;%let points=do(-0.024,6.215,6.239/9);%let weights=repeat(0.1,1,10);%let grid=do(-0.024,6.215,6.239/800);%let parameter={15.03 1.31 530 1587};* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=1;%let const2=1;%let cmerge=12;

Page 181: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

168 Pharmaceutical Statistics Using SAS: A Practical Guide

* G function;%macro deriv(matrix,derivative);

nm=nrow(&matrix);one=j(nm,1,1);dera=(parameter[4]-parameter[3])/(1+(exp(&matrix[,1])/parameter[1])

##parameter[2])##2#(parameter[2]/parameter[1])#(exp(&matrix[,1])/parameter[1])##parameter[2];

derb=-(parameter[4]-parameter[3])/(1+(exp(&matrix[,1])/parameter[1])##parameter[2])##2#(exp(&matrix[,1])/parameter[1])##parameter[2]#log(exp(&matrix[,1])/parameter[1]);

derc=1-1/(1+(exp(&matrix[,1])/parameter[1])##parameter[2]);derd=1/(1+(exp(&matrix[,1])/parameter[1])##parameter[2]);&derivative=dera||derb||derc||derd;

%mend deriv;%OptimalDesign1;

Output from Program 7.3

Initial design

Weight X

0.100 -0.0240.100 0.6690.100 1.3620.100 2.0560.100 2.7490.100 3.4420.100 4.1350.100 4.8290.100 5.5220.100 6.215

Optimal design

Weight X

0.250 -0.0240.250 2.0270.250 3.5400.250 6.215

Determinants of the variance-covariance matrices

INITIAL OPTIMAL

0.000031 0.000015

Variance-covariance matrix, initial design

COL1 COL2 COL3 COL4

0.019 0.000 -0.103 -0.2830.000 0.000 0.027 -0.041

-0.103 0.027 6.745 -3.599-0.283 -0.041 -3.599 12.639

Page 182: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 169

Variance-covariance matrix, optimal design

COL1 COL2 COL3 COL4

0.015 0.000 -0.118 -0.1810.000 0.000 0.013 -0.019-0.118 0.013 5.086 -1.065-0.181 -0.019 -1.065 7.087

Output 7.3 shows that the D-optimal design includes two points on the boundary of thedesign region (x∗

1 = −0.024 and x∗4 = 6.215) and two in the middle of the design region

(x∗2 = 2.027 and x∗

3 = 3.540). The optimal weights are equal to 1/m = 0.25 (compareOutput 7.1); see the left panel in Figure 7.5. As in Examples 1 and 2 given in Section 7.2,the sensitivity function is equal to the number of the model parameters, m = 4, at theoptimal concentrations (see the right panel in Figure 7.5).

Figure 7.5 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivity func-tions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Concentration (log scale)

-0.03 1.02 2.07 3.12 4.17 5.22 6.27

Sen

sitiv

ity

2

3

4

5

6

7

Concentration (log scale)

-0.03 1.02 2.07 3.12 4.17 5.22 6.27

Output 7.3 also helps compare characteristics of the initial and optimal designs. Thedeterminant of the final variance-covariance matrix is 1.5×10−5, compared to the value of3.1×10−5 for the initial design. The variance of individual parameter estimates hasimproved, too. For example, the variance of θ1 has dropped from 0.019 to 0.015. Parameterθ1, or ED50, is usually the most important for practitioners. This example illustrates agood performance of a D-optimal design with respect to other criteria of optimality, likec-criterion for the estimation of θ1.

7.4 Regression Models with Unknown Parameters in theVariance FunctionIn various pharmaceutical applications it is often assumed that the variance of the errorterm, σ2, depends not only on the control variable x, but also on the unknown parameter θ,i.e., σ2

i = S(xi, θ).Consider, for instance, the four-parameter logistic model introduced in Section 7.2. The

variance of the response variable, Y , may depend on its mean, as in the following powermodel (Finney, 1976; Karpinski, 1990; Hedayat et al., 1997):

Var(Y |x, θ) = S(x, θ) ∼ ηδ(x, θ), δ > 0,

Page 183: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

170 Pharmaceutical Statistics Using SAS: A Practical Guide

In cell-based assay studies, the response variable is a cell count and is often assumed tofollow a Poisson distribution. Under this assumption, one can consider a power variancemodel with δ = 1.

Another possible generalization of the nonlinear regression models considered inSection 7.2 is a regression model with multiple correlated responses. Multiple correlatedresponses are encountered in clinical trials with several endpoints or several objectives, e.g.,simultaneous investigation of efficacy and toxicity in dose response studies (Heise andMyers, 1996; Fan and Chaloner, 2001).

7.4.1 General ModelLet the observed response Y be a normally distributed k-dimensional vector with

E[Y |x] = η(x, θ), Var[Y |x] = S(x, θ), (7.11)

where η(x, θ) = (η1(x, θ), . . . , ηk(x, θ))T is a vector of pre-defined functions and S(x, θ) is ak × k positive definite matrix. It can be shown that the Fisher information matrix of asingle observation, μ(x, θ), is the sum of two terms; see Magnus and Neudecker, 1988,Chapter 6, or Muirhead, 1982, Chapter 1:

μ(x, θ) = μR(x, θ) + μV (x, θ), (7.12)

μRij(x, θ) =

∂ηT (x, θ)∂θi

S−1(x, θ)∂η(x, θ)

∂θj, i, j = 1, . . . , m,

μVij(x, θ) =

12tr

[S−1(x, θ)

∂S(x, θ)∂θi

S−1(x, θ)∂S(x, θ)

∂θj

],

where m is the number of unknown parameters. In the univariate case (k = 1), μ(x, θ)permits the following factorization:

μ(x, θ) = gR(x, θ)gTR(x, θ) + gV (x, θ)gT

V (x, θ), (7.13)

where

gR(x, θ) =f(x, θ)√S(x, θ)

, gV (x, θ) =h(x, θ)√2S(x, θ)

,

f(x, θ) is the vector of partial derivatives of η(x, θ) as in (7.10) and h(x, θ) is the vector ofpartial derivatives of S(x, θ) with respect to θ, i.e.,

h(x, θ) =[∂S(x, θ)

∂θ1,

∂S(x, θ)∂θ2

, . . . ,∂S(x, θ)

∂θm

]T

. (7.14)

It is important to note that, since the information matrix μ(x, θ) consists of two terms,its rank may be greater than 1 even in the univariate case. This may lead to the situationwhen the number of support points in the optimal design is less than the number ofunknown parameters, i.e., less than m (see Downing, Fedorov and Leonov (2001) or Fanand Chaloner (2001) for examples). This cannot happen in regression models with a singleresponse variable and a known variance since in this case the information matrix of thedesign would become singular.

Sensitivity Functions

Using classic optimal design techniques, it can be shown that the equivalence theorem(7.6), (7.7) remains valid for multivariate models in which the covariance matrix depends

Page 184: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 171

on unknown parameters (Atkinson and Cook, 1995; Fedorov, Gagnon and Leonov, 2002).In the univariate case, sensitivity functions for the D- and A-optimality criteria can bepresented as the sum of two terms and can be factorized too. For example, the sensitivityfunction for the D-optimality criterion can be written as

ψ(x, ξ, θ) = gR(x, θ)M−1(ξ, θ)gTR(x, θ) + gV (x, θ)M−1(ξ, θ)gT

V (x, θ).

7.4.2 Example 4: A Four-Parameter Logistic Model with a Power VarianceModel

This example is concerned with the computation of the D-optimal design in afour-parameter logistic model that extends the logistic model considered in Example 3 (seeSection 7.3). Assume that Y is a normally distributed response variable with

E[Y |x] = η(x, θ) = θ3 +θ4 − θ3

1 + (x/θ1)θ2,

Var[Y |x] = S(x, θ) = δ1ηδ2(x, θ),

where δ1 and δ2 are positive parameters.Parameters θ1 − θ4 are selected as in Example 3, i.e., θ = (15.03, 1.31, 530, 1587)T and

δ1 = 0.5, δ2 = 1. The combined vector of model parameters is (15.03, 1.31, 530, 1587, 0.5, 1)T .

Computation of the D-Optimal DesignProgram 7.4 computes the D-optimal design for the four-parameter logistic model specifiedabove. This program is conceptually similar to Program 7.3. We need only to make thefollowing changes:

• Add the two variance parameters to the PARAMETER variable.• Change the method of calculating the derivatives since the analytic form of the

derivatives is rather complicated. The %deriv macro computes gR(x, θ) numericallyusing the NLPFDD function. Since the information matrix and sensitivity function alsodepend on gV (x, θ), we introduce a new macro (%derivs) that calculates this functiongV (x, θ) (it is also derived using a numerical approximation).

• Since the information matrix is now the sum of two terms, the %infod macro needs tobe modified as well.

• As in Program 7.3, the value of the merging constant (CMERGE) is increased toimprove the stability of the iterative procedure in this complex optimal design problem.

The complete version of Program 7.4 is provided on the book’s companion Web site.

Program 7.4 D-optimal design for the four-parameter logistic model with a continuous response andunknown parameters in the variance function (Design and algorithm parameters)

* Design parameters;%let points=do(-0.024,6.215,6.239/7);%let weights=repeat(0.125,1,8);%let grid=do(-0.024,6.215,6.239/400);%let parameter={15.03 1.31 530 1587 0.5 1};* Number of parameters;%let paran=6;

Page 185: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

172 Pharmaceutical Statistics Using SAS: A Practical Guide

* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=1;%let const2=1.2;%let cmerge=7.5;%OptimalDesign1;

Output from Program 7.4

Initial design

Weight X

0.125 -0.0240.125 0.8670.125 1.7590.125 2.6500.125 3.5410.125 4.4320.125 5.3240.125 6.215

Optimal design

Weight X

0.294 -0.0240.213 1.9720.236 3.5630.257 6.215

Output 7.4 lists the initial and optimal designs. Other parameters of the two designs,e.g., the variance-covariance matrices and their determinants, can be obtained by printingout the DINITIAL, DOPTIMAL, and DDET data sets. It was emphasized above that, inmodels with unknown parameters in the variance function, the number of support points inoptimal designs can be less than the number of model parameters. Indeed, Output 7.4shows that, although there are two more unknown parameters in this model compared tothe model with a constant variance (Example 3 of Section 7.3), the D-optimal design is stilla four-point design. The weights are no longer equal to 1/4, and are equal to(0.294, 0.213, 0.236, 0.257) for the four design points (−0.024, 1.972, 3.563, 6.215) on thelog-scale.

Figure 7.6 provides a visual comparison of the initial and optimal designs (left panel)and also plots the sensitivity functions associated with the two designs (right panel).

7.5 Models with a Bounded Response (Beta Models)The Beta regression model provides another example where the information matrix can becalculated in a closed form.

7.5.1 General ModelIn clinical trials, investigators often need to deal with ordinal variables containing manycategories. The Beta regression model has been shown to be a good choice to analyze thistype of response; see Wu, Fedorov and Propert (2005).

Page 186: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 173

Figure 7.6 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivity func-tions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Concentration (log scale)

-0.03 1.02 2.07 3.12 4.17 5.22 6.27

Sen

sitiv

ity

4

5

6

7

8

9

Concentration (log scale)

-0.03 1.02 2.07 3.12 4.17 5.22 6.27

In this section we consider an example from a randomized, double-blind,placebo-controlled, parallel group clinical trial. Each patient was randomized to one of fourtreatment arms, with 0, 2, 4, and 8 dose units, respectively. The observations were taken atbaseline and the end of the study at 6 months. The response is a severity measure scorethat varies between 0 and 1 on the normalized scale, with hundreds of levels. Higher scoresindicate higher degrees of severity of the illness.

After investigating the data on the descriptive level, we observed that the median scorehas a slightly decreasing trend when dose increases. But the difference may not be bigenough to be statistically and/or clinically significant. However, the 8 dose units group hasa smaller variance than the other groups. Even among the other three treatment groups,the estimated population variances are different from one group to another. These factsindicate that, although the drug did not reduce the overall mean response, it might beeffective in some subjects.

Let yij denote the response rate from patient j under dose level xi and assume that yij

follows the Beta distribution with parameters p(xi) and q(xi). We can model p(xi) andq(xi) as a function of the dose x:

ln p(x) = αT f(x) and ln q(x) = βT φ(x), (7.15)

Note that

E(yij) =p(xi)

p(xi) + q(xi), Var(yij) =

p(xi)q(xi)(p(xi) + q(xi))2(p(xi) + q(xi) + 1)2 ,

where B(p, q) = Γ(p)Γ(q)/Γ(p + q) and Γ(p) =∫ ∞0 tp−1e−tdt is the Gamma function. Let

θ = (αT , βT ), denote p(xi) and q(xi) as pi and qi, respectively, and consider the Digamma orTrigamma functions (Abramowitz and Stegun, 1972):

ΨΓ(v) =d lnΓ(v)

dv, Ψ′(v) =

dΨΓ(v)dv

.

The information matrix μ(xi, θ) of a single observation at point xi is given by([Ψ′(pi) − Ψ′(pi + qi)]p2

i f(xi)fT (xi) −Ψ′(pi + qi)piqif(xi)φT (xi)−Ψ′(pi + qi)piqiφ(xi)fT (xi) [Ψ′(pi) − Ψ′(pi + qi)]p2

i φ(xi)φT (xi)

). (7.16)

The information matrix of N experiments, with ni independent observations at dose xi,admits presentation (7.2).

Page 187: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

174 Pharmaceutical Statistics Using SAS: A Practical Guide

Here we consider a simple example with α = (α1, α2)T , β = (β1, β2)T , andf(x) = φ(x) = (1, x)T . In that case the individual information matrix may be written asfollows:

μ(xi, θ) =(

[Ψ′(pi) − Ψ′(pi + qi)]p2i −Ψ′(pi + qi)piqi

−Ψ′(pi + qi)piqi [Ψ′(pi) − Ψ′(pi + qi)]p2i

) ⊗(1 xi

xi x2i

), (7.17)

where⊗

is the Kronecker product.Figure 7.7 displays the fitted Beta densities for each treatment group with

θ = (α1, α2, β1, β2)T = (4,−0.49, 3.9, 0.15)T .

The figure shows that, as the dose level increases, the mean response becomes smaller andthus we see evidence of a drug effect. Note also that the variance of the response variableclearly changes with the average response level and the highest dose group has the smallestvariance.

Figure 7.7 Distribution of simulated responses in each treatment group

8 units

4 units

2 units 0 units

Res

pons

e

0

10

20

30

40

Severity measure score

0.0 0.2 0.4 0.6 0.8 1.0

Unlike quantal dose-response models discussed in Section 7.2 or continuous logisticmodels from Sections 7.3–7.4, the presentation (7.16) for the information matrix of theBeta regression model cannot be factorized. As a result, the general formulas (7.6) and(7.7) have to be implemented in this case.

7.5.2 Example 5: Beta Regression ModelTo construct a D-optimal design for the clinical trial example discussed above, we use thefollowing Beta regression model:

ln p(x) = 4 − 0.49x, ln q(x) = 3.9 + 0.15x.

Program 7.5 computes a D-optimal design for the Beta regression model by calling the%OptimalDesign2 macro. The initial design contains 6 equally spaced doses between 0 and8 dose units, i.e., X = [0, 8]. The true value of the parameter vector isθ = (4,−0.49, 3.9, 0.15)T .

The overall optimal design algorithm in this problem remains the same; however, unlikethe previous examples, the information matrix is calculated directly, without using partial

Page 188: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 175

derivatives of the response function. Therefore, in this example (and subsequent examplesin this chapter) the user does not need to specify the %deriv1 or %deriv2 macros. Thedirect calculation of the information matrix relies on the %Info, %infoele and %indinfomacros. The analytical form of the information matrix is specified in the %info macro. The%infoele macro is needed to store the individual information matrices based on the valuesof covariates on the grid. Elements of the information matrices are stored in differentvectors based on the position in the matrix. Note that the information matrix is asymmetric matrix so only the upper (lower) triangular matrix is needed when storing it.For example, the ELEO14 vector contains the elements in the first row, fourth column ofall the information matrices. The %indinfo macro outputs the individual informationmatrix for a given candidate point (note that the MDERIV macro variable is not used inthis program and can be set to any value). The complete SAS code for the updated macrosis provided on the book’s companion Web site.

Program 7.5 D-optimal design for the beta regression model (Design and algorithm parameters)

* Design parameters;%let points=do(0,8,8/5);%let weights=repeat(1/6,1,6);%let grid=do(0,8,8/200);%let parameter={4 -0.49 3.9 0.15};* Number of parameters;%let paran=4;* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=2;%let const2=1;%let cmerge=5;%OptimalDesign2;

Output from Program 7.5

Initial design

Weight X

0.167 0.0000.167 1.6000.167 3.2000.167 4.8000.167 6.4000.167 8.000

Optimal design

Weight X

0.491 0.0000.071 4.2800.438 8.000

Output 7.5 shows the 6-point initial and 3-point optimal designs. Note that most of theweight in this D-optimal design is assigned to the two points on the boundaries of the doserange (placebo and 8 dose units). The middle point receives very little weight (only 7% of

Page 189: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

176 Pharmaceutical Statistics Using SAS: A Practical Guide

the patients will be allocated to this dose). Figure 7.8 displays the initial and optimaldesigns and their sensitivity functions.

Figure 7.8 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivity func-tions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Dose

0 2 4 6 8

Sen

sitiv

ity

2

4

6

8

Dose

0 2 4 6 8

7.6 Models with a Bounded Response (Logit Link)The Beta regression model takes care of the bounded variable naturally (by definition). Inthis section we introduce an alternative way to model the dose-response trial example fromSection 7.5, using the linear regression model framework. After a simple transformation,the responses are not bounded and can be treated as normally distributed variables.Furthermore, to address the relationship between variance and dose level, one can modelthe variance as a function of the dose.

7.6.1 One-Dimensional CaseThe model with a single drug can be written as

lnB − Y

Y − A= θ1 + θ2x + σ2(x, θ)ε,

where the variance function is defined as follows

σ2(x, θ) = exp(θ3 + θ4x).

Further, Y is the response varying from A to B, x denotes the dose, and the error term isnormally distributed, i.e., ε ∼ N(0, 1).

In this example we take A = 0 and B = 1. Note that this model provides anotherexample of models with unknown parameters in the variance function (compare with(7.11)). Thus formulas (7.13) and (7.14) can be applied here, with

f(x, θ) = (1, x, 0, 0)T , h(x, θ) = (0, 0, 1/√

2, x/√

2)T .

The information matrix of a single observation is

μ(x, θ) =( 1

exp(θ3+θ4x) 00 0.5

) ⊗(1 xx x2

).

It follows from this equation that an optimal design will depend only on the values of θ3and θ4.

Page 190: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 177

7.6.2 Example 6: A Bounded-Response Model with a Logit Link and UnknownParameters in the Variance Function

Program 7.6 calls the %OptimalDesign2 macro to find the D-optimal design for thebounded-response model defined earlier in this subsection. We need to change the %infoelemacro that computes the information matrix and remove the %devpsi macro. The othermacros do not change compared to Example 5. The complete SAS code for Program 7.6can be found on the book’s companion Web site.

Note that, as in Program 7.5, the initial design includes six equally spaced points in thedose region X = [0, 8]. The parameter verctor is given by θ = (1,−0.5, 1,−0.6)T and thefitted normal densities for each treatment group are displayed in Figure 7.9 (the densitiesare shown on a normal scale, i.e., ln(1 − y)/y). As in Figure 7.7, we see that, as the doselevel increases, the mean response (disease severity) decreases and the variance decreases aswell.

Figure 7.9 Distribution of the response variable in each treatment group

8 units

4 units2 units

0 units

Res

pons

e

0

2

4

Normal score

-5 -4 -3 -2 -1 0 1 2 3 4 5

Program 7.6 D-optimal design for the bounded-response model with a logit link and unknown parameters inthe variance function (Design and algorithm parameters)

* Design parameters;%let points=do(0,8,8/5);%let weights=repeat(1/6,1,6);%let grid=do(0,8,8/400);%let parameter={-1 0.5 -1 -0.6};* Number of parameters;%let paran=4;* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=1;%let const2=1;%let cmerge=3;%OptimalDesign2;

Page 191: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

178 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 7.6

Initial design

Weight X

0.167 0.0000.167 1.6000.167 3.2000.167 4.8000.167 6.4000.167 8.000

Optimal design

Weight X

0.307 0.0000.228 4.7200.465 8.000

Output 7.6 displays the initial and D-optimal designs. The optimal design is athree-point design with two points on the boundaries and one design point inside thedesign region. Comparing this optimal design with the design shown in Output 7.5, it iseasy to see that the middle dose (4.720) is not much different from the optimal middle dosecomputed from the Beta regression model (4.280); however, it receives a much greaterweight. The initial and optimal designs as well as the associated sensitivity functions aredepicted in Figure 7.10.

Figure 7.10 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivityfunctions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Dose

0 2 4 6 8

Sen

sitiv

ity

2

4

6

8

Dose

0 2 4 6 8

7.6.3 Models with a Bounded Response (Combination of Two Drugs)Under the same dose-response set-up as in Section 7.5, we now consider the case of atwo-drug combination: Drug A and Drug B. The daily dose of each drug ranges from 0.1mg to 10 mg. Due to safety restrictions in drug development, often it is not safe to assignpatients to the treatment with both drugs at the highest dose. In this example, we requirethat the total dose of the two drugs cannot exceed 10.1 mg. Note that the resulting design

Page 192: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 179

region is not square, it is a triangle.Let x and z denote the daily doses of Drugs A and B, respectively. The corresponding

model is

ln1 − y

y= θ1 + θ2x + θ3z + θ4xz + θ5x

2 + θ6z2 + σ2(x, θ, z)ε,

where the variance function is given by

σ2(x, θ, z) = exp(θ7 + θ8x + θ9z).

It can be shown that the information matrix of a single observation is

μ(xi, zj , θ) =(

exp(−θ7 − θ8xi − θ9zj)f1fT1 0

0 0.5f2fT2

),

where i = 1, . . . , n, j = 1, . . . , n,

f1 = (1, xi, zj , xi, zj , x2i , z2

j )T , f2 = (1, xi, zj)T .

Here n is the number of possible doses of each drug. Note that, similar to theone-dimensional case considered in Example 6, the optimal design depends only on theparameters of the variance function, i.e., θ7, θ8 and θ9.

7.6.4 Example 7: Two Design Variables (Two Drugs)Program 7.7 computes the D-optimal design for the introduced model. This program callsthe %OptimalDesign3 macro that supports a two-dimensional version of the optimal designalgorithm defined in Section 7.1.5. The design parameters are specified in the%DesignParameters macro and then they are passed to the %OptimalDesign3 macro. Theinitial design (POINTS and WEIGHTS variables) includes ten equally weighted pointsthat are evenly distributed across the triangular design region:

0.1 ≤ x ≤ 10, 0.1 ≤ z ≤ 10, x + z ≤ 10.1.

Program 7.7 D-optimal design for the bounded-response model with two design variables (Design andalgorithm parameters)

%macro DesignParameters;* Initial design: Design points;* Four equally spaced doses for each drug;dosen=4;* Total number of points;dosem=dosen*(dosen+1)/2;points=j(dosem,2,0);k=0;do i=1 to dosen;

do j=1 to dosen-i+1;k=k+1;points[k,1]=0.1+(i-1)*9.9/(dosen-1);points[k,2]=0.1+(j-1)*9.9/(dosen-1);

end;end;* Initial design: Weights;* Equally weighted points;weights=t(repeat(1/10,1,10));

Page 193: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

180 Pharmaceutical Statistics Using SAS: A Practical Guide

* Grid;* 26 equally spaced points on each axis;gridn=26;gridm=gridn*(gridn+1)/2;grid=j(gridm,2,0);k=0;do i=1 to gridn;

do j=1 to gridn-i+1;k=k+1;grid[k,1]=0.1+(i-1)*9.9/(gridn-1);grid[k,2]=0.1+(j-1)*9.9/(gridn-1);

end;end;* Parameter values;para={1 1 1 1 1 1 1 0.6 0.4};

%mend DesignParameters;* Number of parameters;%let paran=9;* Algorithm parameters;%let convc=1e-9;%let maximit=500;%let const1=1;%let const2=1;%let cmerge=3;%OptimalDesign3;

Output from Program 7.7

Initial design

Drug DrugWeight A B

0.10 0.10 0.100.10 0.10 3.400.10 0.10 6.700.10 0.10 10.00.10 3.40 0.100.10 3.40 3.400.10 3.40 6.700.10 6.70 0.100.10 6.70 3.400.10 10.0 0.10

Optimal design

Drug DrugWeight A B

0.17 0.10 0.100.14 0.10 2.870.20 0.10 10.00.14 2.48 0.100.14 3.66 5.640.21 10.0 0.10

Page 194: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 181

Figure 7.11 Left panel: two-dimensional grid for the optimal design problem involving a combination therapy(dots) and initial design (open circles). Right panel: optimal design (closed circles)

Dos

e (D

rug

A)

0

2

4

6

8

10

Dose (Drug B)

0 2 4 6 8 10

Dos

e (D

rug

A)

0

2

4

6

8

10

Dose (Drug B)

0 2 4 6 8 10

The grid (GRID variable) consists of 351 points in the design region. Figure 7.11 (leftpanel) depicts the selected grid (dots) and initial design (open circles). The parametervector (PARAMETER variable) is given by

θ = (1, 1, 1, 1, 1, 1, 1, 0.6, 0.4).

The information matrix and sensitivity function are defined using the %infoele,%indinfo, and %info macros. The complete version of Program 7.7 is given on the book’scompanion Web site.

Output 7.7 displays the initial and D-optimal designs. The right panel of Figure 7.11provides a graphical summary of the optimal design (closed circles). It is worth noting thatfive of the six points in the optimal design lie on the boundary of the design region. Thecorresponding combination therapies involve the minimum dose (0.1 mg) of at least one ofthe two drugs. There is only one “non-trivial” combination with 3.7 mg of Drug A and 5.6mg of Drug B.

7.7 Bivariate Probit Models for Correlated Binary ResponsesIn clinical trial analysis, the toxicity and efficacy responses usually occur together and itmay be useful to assess them together. However, in practice the assessment of toxicity andefficacy is sometimes separated. For example, determining the maximum tolerated dose(MTD) is based on toxicity alone and then efficacy is evaluated in Phase II trials over thepredetermined dose range. Obviously, the fact that the two responses from the samepatient are correlated will introduce complexity into the analysis. But if we study theseoutcomes simultaneously, more information will be gained for future trials and treatmenteffects will be understood more thoroughly. In drug-response relationship, the correlationbetween efficacy and toxicity can be negative or positive depending on the therapeuticalarea. Two commonly used models, the Gumbel model (Kotz et al., 2000) and the bivariatebinary Cox model (Cox, 1970), have been introduced to incorporate the two dependentoutcomes, toxicity and efficacy, when both of them are dichotomous. In those two models,we need to model the probabilities of different outcome combinations separately. Whenboth outcomes are binary, the total number of combinations is four, but when outcomescontain more than two categories, the number of unknown parameters may increasedramatically. Here we propose a bivariate probit model which incorporates the correlatedresponses naturally via the correlation structure of the underlying bivariate normal

Page 195: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

182 Pharmaceutical Statistics Using SAS: A Practical Guide

distribution; see Lesaffre and Molenberghs (1991). When the number of responses is morethan two, the multivariate probit model may be used in a similar fashion.

Let Y = {0 or 1} denote the efficacy response and U = {0 or 1} denote the toxicityresponse in a clinical trial. Here 0 indicates no response and 1 indicates a response. Let ddenote the dose of a drug and

pyu(x) = P (Y = y, U = u|d = x), y, u = 0 or 1.

Assume that Z1 and Z2 follow bivariate normal distribution with zero mean andvariance-covariance matrix

Σ =(

1 ρρ 1

),

where ρ may be interpreted as the correlation measure between toxicity and efficacy for thesame patient. This set-up may be viewed as a standardization of the observed responses Yand U since under the natural scale the mean and variance vary from study to study. Aftersimple transformation, the correlated responses follow the standard bivariate normaldistribution,

p11 = F (θT1 f1, θ

T2 f2) =

∫ θT1 f1

−∞

∫ θT2 f2

−∞

12π|Σ|1/2 exp(−1

2ZT Σ−1Z)dz1dz2,

where θ1, θ2 are unknown parameters and f1(x) and f2(x) contain the covariates of interest.In this section, we will study a simple linear model defined as follows (refer to Fedorov,Dragalin, and Wu, 2006).

θT1 f1 = θ11 + θ12x, θT

2 f2 = θ21 + θ22x.

In this case, the efficacy and toxicity response rates (p1. and p.1, respectively) can beexpressed as the marginals of the bivariate normal distribution,

p1. = Φ(θT1 f1), p.1 = Φ(θT

2 f2),

where Φ(z) is the cumulative distribution function of the standard normal distribution.Note that p11, p1., and p.1 uniquely define the joint distribution of Y and U , i.e.,p10 = p1. − p11, p01 = p.1 − p11 and p00 = 1 − p1. − p.1 + p11.

Assume that the {yi, ui}’s are independent for different i’s. Then the likelihood functionfor {Y,U} is given by

L(Y,U |θ) =N∏

i=1

yiui log p11 + yi(1 − ui) log p01 + (1 − yi)(1 − ui) log p00.

The information matrix of a single observation is[[C1C2]

⊗f

ϕ2 −ϕ2 −ϕ2

](P − ppT )−1

[[C1C2]

⊗f

ϕ2 −ϕ2 −ϕ2

]T

,

where

C1 =(

ϕ(θT1 f) 00 ϕ(θT

2 f)

), C2 =

(F (u1) 1 − F (u1) −F (u1)F (u2) −F (u2) 1 − F (u2)

),

u1 =θT2 f2 − ρθT

1 f1√1 − ρ2

, u2 =θT1 f1 − ρθT

2 f2√1 − ρ2

,

P =

⎛⎝ p11 0 00 p10 00 0 p01

⎞⎠ , p = (p11 p10 p01)T ,

ϕ(v) denotes the probability density function of the standard normal distribution,ϕ2 = f(θT

1 f, θT2 f, ρ) denotes the probability density function of bivariate normal distribution

with mean θT1 f1 and θT

2 f2, variance 1 and correlation coefficient ρ.

Page 196: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 183

7.7.1 Example 8: A Bivariate Probit Model for Correlated Binary Responses(Efficacy and Toxicity)

Consider the problem of constructing the D-optimal design for the following model:

θT1 f1 = −0.9 + 10x, θT

2 f2 = −1.2 + 1.6x, ρ = 0.5.

Here x denotes the dose of the experimental drug with the dose range given by X = [0, 1].Figure 7.12 depicts the response probabilities for

θ = (−0.9, 10,−1.2, 1.6)T and ρ = 0.5.

This setting represents a scenario which is often encountered in clinical trial applications.The probabilities of efficacy and toxicity responses, p.1 and p1., are both increasing as thedose increases. On the other hand, probability of “positive” response p10 (i.e., probability ofpositive efficacy and no toxicity) increases at the beginning. Then, at a certain dose level itbegins to decrease (the probability of having a positive efficacy response without any sideeffects is low at high dose levels).

Figure 7.12 Bivariate probit model, probability of positive response (solid line), probability of efficacy response(dashed line), and probability of toxicity response (dotted line)

Pro

babi

lity

of r

espo

nse

0.0

0.2

0.4

0.6

0.8

1.0

Dose on a log scale

0.0 0.2 0.4 0.6 0.8 1.0

Program 7.8 constructs the D-optimal design for the described bivariate probit modelby calling the %OptimalDesign4 macro. The initial design includes four equally spaceddoses in the dose range X = [0, 1] (POINTS variable), and the doses are assumed to beequally weighted (WEIGHTS variable). The grid consists of 401 equally spaced points(GRID variable), and the PARAMETER variable specifies the true values of θ and ρ. Theinformation matrix and sensitivity function are computed using the %infoele, %info, and%infod macros. The complete version of Program 7.8 is provided on the book’s companionWeb site.

Page 197: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

184 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 7.8 D-optimal design for the bivariate probit model with correlated binary responses (Design andalgorithm parameters)

* Design parameters;%let points={0 0.333 0.667 1};%let weights={0.25 0.25 0.25 0.25};%let grid=do(0,1,1/400);%let parameter={-0.9 10 -1.2 1.6 0.5};* Number of parameters;%let paran=5;* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=1;%let const2=1;%let cmerge=5;%OptimalDesign4;

Output from Program 7.8

Initial design

Weight X

0.250 0.0000.250 0.3330.250 0.6670.250 1.000

Optimal design

Weight X

0.451 0.0000.356 0.1800.194 1.000

Output 7.8 displays the initial and D-optimal designs. The locally D-optimal design is athree-point design with unequal weights. Two optimal doses are on the boundaries of thedose range (x∗

1 = 0, x∗2 = 1) and the other optimal dose lies in the middle (x∗

3 = 0.18).Almost half of the patients are assigned to the lowest dose and about 20% of patients areassigned to the highest dose. Figure 7.13 depicts the initial and optimal designs as well asthe sensitivity funtions.

7.8 Pharmacokinetic Models with Multiple Measurements per PatientIn this section we discuss a clinical pharmacokinetic (PK) study where multiple bloodsamples are taken for each enrolled patient. This setup leads to nonlinear mixed effectsregression models with multiple responses.

7.8.1 Two-Compartment Pharmacokinetic ModelConsider a clinical trial in which the drug was administered as a bolus input D0 at thebeginning of the study (at time x = 0) and then bolus inputs Di were administered at 12-or 24-hour intervals until 72 hours after the first dose. To describe the concentration of

Page 198: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 185

Figure 7.13 Left panel: Initial (open circles) and optimal (closed circles) designs. Right panel: Sensitivityfunctions for the initial (dashed curve) and optimal (solid curve) designs.

Wei

ght

0.0

0.2

0.4

0.6

Dose

0.0 0.2 0.4 0.6 0.8 1.0

Sen

sitiv

ity

0

4

8

12

16

Dose

0.0 0.2 0.4 0.6 0.8 1.0

drug at time x, a two-compartment model was used with standard parameterization(volume of distribution V , transfer rate constants K12, K21, and K10), see Figure 7.14.

Figure 7.14 Diagram of the two-compartment model

K12

10K

1 2D

21K

Measurements of drug concentration yij were taken from the central compartment(compartment 1 in Fig. 7.14) at times xj for each patient,

yij =g1(xj)

Vi+ zij , i = 1, . . . , N, j = 1, . . . , k, (7.18)

g1(xj) = g1(xj , γi) is the amount of the drug in the central compartment at time xj forpatient i, γi = (Vi, K12,i, K21,i, K10,i) are individual PK parameters of patient i, and zij aremeasurement errors. The g1 function and population model are discussed in more detailbelow.

In the trial under consideration, k = 16 samples were taken from each patient at timesxj ∈ X ,

xj ∈ X = {5, 15, 30, 45 min; 1, 2, 3, 4, 5, 6, 12, 24, 36, 48, 72, 144 h}.

Obviously, each extra sample provides additional information and increases the precision ofparameter estimates. However, the number of samples that may be drawn from eachpatient is restricted because of blood volume limitations and other logistical and ethicalreasons. Moreover, the analysis of each sample is associated with monetary costs.Therefore, it is reasonable to take the cost of drawing samples into account. IfX = (x1, ..., xk) is a collection of sampling times for a particular patient, we can considerthe cost of the sampling sequence X (denoted by c(X)).

Page 199: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

186 Pharmaceutical Statistics Using SAS: A Practical Guide

In this section we will consider the following questions:

1. Given restrictions on the number of samples for each patient, what are the optimalsampling times (i.e., how many samples and at which times)?

2. If not all 16 samples are taken, what would be the loss of information/precision?3. If costs are taken into account, what is the optimal sampling scheme? Cost-based

designs will be discussed in detail in Section 7.9.

By an optimal sampling scheme we mean a sequence which allows us to obtain the bestprecision of parameter estimates in terms of their variance-covariance matrix and theselected optimality criterion. First, we need to define the regression model with multipleresponses, see (7.11). In this and next sections, X will denote a k-dimensional vector ofsampling times and Y will be a k-dimensional vector of measurements at times X.

7.8.2 Regression ModelA two-compartment model described by the following system of linear differentialequations was used to model the data:{

q1(x) = −(K12 + K10

)q1(x) +K21q2(x)

q2(x) = +K12q1(x) −K21q2(x), (7.19)

for x ∈ [ti, ti+1) with initial conditions

q1(ti) = q1(ti − 0) + Di, q1(0) = D0, q2(0) = 0,

where q1(x) and q2(x) are amounts of the drug at time x in the central and peripheralcompartments, respectively; ti is a time of i-th bolus input, t0=0; Di is the amount of thedrug administered at ti.

The solution of the system (7.19) is a sum of exponential functions which depend onparameters (Gibaldi and Perrier, 1982, Appendix B):

γ = (V,K12, K21, K10)T , q(x) = [q1(x, γ), q2(x, γ)].

In population modeling it is often assumed that the γi parameters for patient i areindependently sampled from the normal population with

E(γi) = γ0,Var(γi) = Λ, i = 1, . . . , N, (7.20)

where the mγ-dimensional vector γ0 and (mγ × mγ) matrix Λ are usually referred to as the“population”, or “global”, parameters; see Gagnon and Leonov (2005) for a discussion onother population distributions.

To fit the model, it was assumed that the variance of the error term zij in (7.18)depends on the model parameters through the mean response,

zij = ε1,ij + ε2,ijq1(xj)

Vi, (7.21)

where ε1,ij and ε2,ij are independent, identically distributed random variables with zeromean and variance σ2

1 and σ22, respectively.

Let θ = (γ0,Λ, σ21 , σ

22)T denote a combined vector of model parameters and ki denote the

number of samples taken for patient i. Let

Xi = (x1i, x2i, . . . , xki,i)T and Yi = (y1i, y2i, . . . , yki,i)

T

Page 200: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 187

be sampling times and measured concentrations, respectively, for patient i. Further,η(xji, γi) = q1(xji, γi)/Vi, η(xji, θ) = q1(xji, γ

0)/V and

η(Xi, θ) = [η(x1i, θ), . . . , η(xki,i, θ)]T

,

where j = 1, 2, . . . , ki.If F is a (ki × mγ) matrix of partial derivatives of function η(Xi, θ) with respect to

parameters γ0,

F = F (Xi, γ0) =

[∂η(Xi, θ)

∂γα

]∣∣∣∣γ=γ0

,

then, using the first order approximation together with (7.20) and (7.21), one obtains thefollowing approximation of the variance-covariance matrix S(Xi, θ) for Yi,

S(Xi, θ) FΛF T + σ22Diag[η(Xi, θ)ηT (Xi, θ) + FΛF T ] + σ2

1Iki , (7.22)

where Diag(A) denotes a diagonal matrix with elements aii on the diagonal; see alsoFedorov, Gagnon and Leonov (2002; Section 4). If the γ parameters are assumed to belog-normally distributed, then Λ on the right-hand side of (7.22) has to be replaced with

Λ1 = Diag(γ0)ΛDiag(γ0),

for details, see Gagnon and Leonov (2005). Therefore, for any sequence Xi, the responsevector η(Xi, θ) and variance-covariance matrix S(Xi, θ) are defined, and we can use generalformula (7.12) to calculate the information matrix μ(Xi, θ).

7.8.3 Example 9: Pharmacokinetic Model without Cost ConstraintsConsider a problem of selecting an optimal sampling scheme with a single bolus input attime x = 0. SAS code for generating optimal designs for the general case of multiple bolusinputs is available (Gagnon and Leonov, 2005), but the complexity of this code is beyondthe scope of the chapter.

To fit the data, the K12 and K21 parameters were treated as constants (K12 = 0.400,K21 = 0.345), i.e., they were not accounted for while calculating the information matrixμ(x, θ). Therefore, the combined vector of unknown parameters θ was defined as

θ =(γ0

1 , γ02 ;λ11, λ22, λ12;σ2

1 , σ22),

where

γ01 = CL, γ0

2 = V, λ11 = Var(CL), λ12 = Cov(CL, V ), λ22 = Var(V ),

CL is the plasma clearance, and K10 = CL/V . The total number of unknown parameters ism = 7.

To construct locally optimal designs, we obtained the parameter estimates θ using theNONMEM software (Beal and Sheiner, 1992) based on 16 samples (from the design regionX ) for each of 27 subjects. The resulting estimate is given by

θ = (0.211, 5.50; 0.0365, 0.0949, 0.0443; 8060, 0.0213)T .

Drug concentrations were expressed in μg/L, elimination rate constant K10 in L/h andvolume V in L.

The PK modeling problem considered in this section greatly increases the complexity ofthe SAS implementation of D-optimal designs. Program 7.9 relies on a series of SASmacros and SAS/IML modules to solve the optimal selection problem formulated earlier inthis section. Referring to Section 7.1.7, Components 1, 2, and 3 of the general optimaldesign algorithm require extensive changes. These changes are summarized below.

Page 201: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

188 Pharmaceutical Statistics Using SAS: A Practical Guide

• In order to generate the grid, the set of sampling times X is defined in the CAND dataset and the number of time points r in the final design is specified in the KS data set (inthis case, we are looking for 8-point designs). Note that r ranges between 1 and k, wherek is the total number of sampling times (k = 16 in this case). Any number of sets of sizer can be applied to the optimization algorithm; however, for designs with the standardinformation matrix normalization, the design with the largest r will be D-optimal. Thiswill not (necessarily) be true for cost-normalized designs (see Section 7.9).

• The problem of selecting candidate points is a C(k, r) problem, where C(k, r) is abinomial coefficient, C(k, r) = k!/[r!(k − r)!]. Sets of size r candidate points can becomevery large. For example, for k = 16 and r = 8, the number of possible 8-point samplingschemes is C(16, 8) = 12, 870 which means that 12,870 information matrices need to beobtained. Note that in this example m = 7 and thus each information matrix is a 7 × 7matrix. In order to store the information matrices for all possible 8-point designs, SASneeds to store 12, 870 × 7 = 90, 090 rows (and 7 columns) of data. Hence the datastorage requirements for large designs can become formidable. Once the CAND and KSdata sets are specified, Program 7.9 will generate the set of possible candidate pointsusing the SAS/IML module TS. Also, the initial design is specified in the SAMPLE dataset (note that all points in the initial design must be elements of the design region Xand must be of length r, as specified in the KS dataset).

• The set of parameters has been defined as θ = (γ01 , γ

02 , λ11, λ22, λ12, σ

21 , σ

22)T . Since we are

dealing with a mixed effects model, the γ and λ parameters in the vector θ must beidentified. This is accomplished by ordering the parameters in a specific way. In general,the parameters in θ must be entered as follows: vector γ, vector λ, and residualvariance(s). The number of components in γ is defined by the NF macro variable. Theordering of the random effects (the λ’s) is also highly important. Parameters along themain diagonal of Λ should be entered first: λ11, λ22. The off-diagonal element (λ12)should be entered next. The Λ matrix is set up using the SAS/IML module LSETUP. Inthe general case, when Λ is a b × b matrix, the components of Λ need to be ordered asfollows: diagonal elements, λ11, λ22, . . . , λbb, followed by off-diagonal elementsλ12, λ13, . . . , λ1b, λ23, λ24, λ2b, . . ., etc.

• The PK function, η(x, θ), derived from (7.19), is specified using the SAS/IML moduleFPARA. The ordering of the γ’s in the vector of parameters must match the ordering inthe FPARA module. As shown in Program 7.9, CL (plasma clearance) should beentered into θ first, and V (volume of distribution) should be entered second. In thecurrent version, the equations in (7.19) must be solved, and the solution must beentered in the FPARA module.

• A computationally difficult issue is the calculation of derivatives for (7.12). Note thatfirst- and second-order partial derivatives are required; see equations (7.12), (7.22). Inthe previous examples derivatives were obtained by either entering the analytical formor by using the NLPFDD function in SAS/IML. Here, we find it more practical tocalculate the derivatives numerically using the following approximation

∂η(x, θ) =η(x, θ + h) − η(x, θ − h)

2h,

where the value of h is a macro variable and is entered by the user (h = 0.001 in thisexample). The information matrix from (7.12) is computed via the SAS/IML moduleMOLD, which calls six other modules (HIGHERD, HIGHERM, FPLUS, FMINUS,FTHETA and JACOB2).

The complete SAS code for this example is provided on the book’s companion Web site.

Page 202: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 189

Program 7.9 D-optimal design for the pharmacokinetic model (Design and algorithm parameters)

* Design parameters;%let h=0.001; * Delta for finite difference derivative approximation;%let paran=7; * Number of parameters in the model;%let nf=2; * Number of fixed effect parameters;%let cost=1; * Cost function (1, no cost function,

2, user-specified function);* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=2;%let const2=1;%let cmerge=5;* PK parameters;data para;

input CL V vCL vV covCLV m s;datalines;0.211 5.50 0.0365 0.0949 0.0443 0.0213 8060;

* All candidate points;data cand;

input x @@;datalines;0.083 0.25 0.5 0.75 1 2 3 4 5 6 12 24 36 48 72 144;

* Number of time points in the final design;data ks;

input r @@;datalines;8;

* Initial design;data sample;

input x1 x2 x3 x4 x5 x6 x7 x8 w @@;datalines;0.083 0.5 1 4 12 24 72 144 1.0;run;

Page 203: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

190 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 7.9

Determinant of the covariance matrix D (initial design)

IDED

0.0000136

Determinant of the covariance matrix D (final design)

DETD

8.4959E-6

Optimal design

COL1 COL2 COL3 COL4 COL5 COL6 COL7 COL8

0.083 0.25 0.5 0.75 36 48 72 144

Output 7.9 lists the optimal sampling times included in a D-optimal 8-point design (5min, 15 min, 30 min, 45 min, 36 h, 48 h, 72 h and 144 h). The optimal design differs fromthe initial design in that it puts more weight on earlier time points; e.g., the optimal designincludes the 15-min and 45-min which were not in the initial design. Note also that anapplication of the D-optimal algorithm reduced the determinant of the optimalvariance-covariance matrix to 8.49 × 10−6 (from 1.36 × 10−5).

7.9 Models with Cost ConstraintsTraditionally, when normalized designs are discussed, the normalization factor is equal tothe number of experiments N ; see (7.3). In this section we will consider cost-normalizeddesigns. Each measurement at point Xi is assumed to be associated with a cost c(Xi), anda restriction on the total cost is given by

n∑i=1

nic(Xi) ≤ C.

In this case it is quite natural to normalize the information matrix by the total cost C andintroduce

MC(ξ, θ) =MN(θ)

C=

∑i

wiμ(Xi, θ), with wi =nic(Xi)

C, μ(X, θ) =

μ(X, θ)c(X)

. (7.23)

Once the cost function c(X) is defined, we can introduce a cost-based designξC = {wi, Xi} and use exactly the same techniques of constructing continuous optimaldesigns for various optimality criteria as described in the previous sections,

ξ∗ = arg minξ

Ψ[DC(ξ, θ)], where DC(ξ, θ) = M−1C (ξ, θ) and ξ = {wi, Xi}.

As usual, to obtain counts ni, values ni = wiC/c(Xi) have to be rounded to the nearestintegers ni subject to

∑i nic(Xi) ≤ C.

We believe that the introduction of cost constraints and the alternative normalization(7.23) makes the construction of optimal designs for models with multiple responses moresound. It allows for a meaningful comparison of “points” X with distinct number ofresponses.

Page 204: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 191

7.9.1 Example 10: Pharmacokinetic Model with Cost ConstraintsThe example considered in this section is the example discussed in Section 7.8.3 with costfunctions added. Program 7.10 computes a D-optimal design by invoking the same SASmacros and SAS/IML modules as Program 7.9. The computational tools for calculating thecost-normalized information matrices are built into the macros and modules; we need onlyto specify the cost function and set the COST variable to 2 (to indicate that a user-definedcost function will be provided).

The cost function must be entered by the user into the SAS/IML module MCOST. InProgram 7.10, we select a linear cost function with two components, the CV variable isassociated with an overall cost of the study (i.e., the cost of a patient visit), and CS is thecost of obtaining/analyzing a single sample:

* Cost module;start mcost(t);

* t is vector sampling times;Cv = 1;Cs = 0.3;C = Cv + sum(Cs[1:nrow(t[loc(t>0)])]);return(C);

finish(mcost);

In general, the cost function can have almost any form.The design region is defined as follows

X1 ={X = (ti1 , . . . , tir), tij ∈ X , j = 1, 2, . . . , r, 3 ≤ r ≤ 5

},

that is, we allow any combination of r sampling times for each patient from the originalsequence X , 3 ≤ r ≤ 5. The values of r are specified in the KS data set. Note that a total of6,748 information matrices (47,236 rows and 7 columns of data) are calculated and storedin this example.

Program 7.10 D-optimal design for the pharmacokinetic model with cost constraints

* Design parameters;%let h=0.001; * Delta for finite difference derivative approximation;%let paran=7; * Number of parameters in the model;%let nf=2; * Number of fixed effect parameters;%let cost=2; * Cost function (1, no cost function, 2, user-specified function);* Algorithm parameters;%let convc=1e-9;%let maximit=1000;%let const1=2;%let const2=1;%let cmerge=5;* PK parameters;data para;

input CL V vCL vV covCLV m s;datalines;0.211 5.50 0.0365 0.0949 0.0443 0.0213 8060;

* All candidate points;data cand;

input x @@;datalines;0.083 0.25 0.5 0.75 1 2 3 4 5 6 12 24 36 48 72 144;

Page 205: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

192 Pharmaceutical Statistics Using SAS: A Practical Guide

* Number of time points in the final design;data ks;

input r @@;datalines;3 4 5;

* Initial design;data sample;

input x1 x2 x3 x4 x5 w @@;datalines;0.083 0.5 4 24 144 1.0;run;

Output from Program 7.10

Determinant of the covariance matrix D (initial design)

IDED

0.231426

Determinant of the covariance matrix D (optimal design)

DETD

0.0209139

Optimal design

Obs COL1 COL2 COL3 COL4 COL5

1 0.083 0.25 48 72 1442 0.083 0.25 72 144

Optimal weights

Obs W

1 0.591252 0.40875

Output 7.10 displays the D-optimal design based on the linear cost funtion defined inthe MCOST module. The D-optimal design is a collection of two sampling sequences,

ξ∗ = {X∗1 = (5, 15 min, 48, 72, 144 h), X∗2 = (5, 15 min, 72, 144 h)},

with weights w1 = 0.59 and w2 = 0.41, respectively. This example shows that once costs aretaken into account, sampling sequences with a smaller number of samples may becomeoptimal.

7.10 SummaryDesign optimization is a critical aspect of experimentation; this is particularly true inpharmaceutical applications. There are many barriers to routine application of optimaldesign theory. Among these is the lack of software. For the mathematically simplest cases,

Page 206: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 193

namely linear models where the parameters are independent of the variance function, manysoftware packages are available for finding optimal designs (see Section 7.1.6). However, asmodels increase in complexity, the availability of software for solving optimal designproblems drops off dramatically and pharmaceutical researchers often find it frustrating.Some academic labs have created software that supports selected optimal designs (seeSection 7.1.6) but a general software package accessible to researchers in thepharmaceutical industry is not currently available. To this end, we have created a series ofpowerful modules that are based on SAS software for constructing optimal experimentaldesigns for a large number of popular non-linear models. Although we focus on D-optimaldesigns, the same algorithm can be used for constructing other types of optimal designs, inparticular A-optimal designs after minor changes.

In this chapter, we provide a brief introduction to optimal design theory, and we alsodiscuss optimal experimental designs for nonlinear models arising in variouspharmaceutical applications. We provide several examples, and the SAS code for executingthese examples is included. The examples increase in complexity as the chapter movesforward. The first example generates D-optimal designs for quantal models; subsequentexamples, in order, generate D-optimal designs for continuous logistic models, logisticregression models with unknown parameters in the variance function, the beta regressionmodel, models with binary response, bivariate probit models for correlated binaryresponses, and pharmacokinetic models with multiple measurements per patient, with orwithout cost constraints. The most important hurdle in contructing these designs is thecomputation of the Fisher information matrix, and storage/retrieval of these matrices andassociated design points. As designs increase in complexity, computation time is increasedin order to handle the necessary calculations and storage/retrieval.

ReferencesAbramowitz, M., Stegun, I. A. (1972). Handbook of Mathematical Functions with Formulas, Graphs

and Mathematical Tables. 9th edition. New York: Dover.Agresti, A. (1990). Categorical Data Analysis. New York: Wiley.Anderson, J.A. (1984). “Regression and ordered categorical variables.” Journal of Royal Statistical

Society. Series B. 46, 1–30.Ashby, D., Pocock, S.J., Shaper, A.G. (1986). “Ordered polytomous regression: An example relating

serum biochemistry and haematology to alcohol consumption.” Applied Statistics. 35, 289–301.Atkinson, A.C., Cook, R.D. (1995). “D-optimum designs for heteroscedastic linear models.” Journal

of the American Statistical Association. 90, 204–212.Atkinson, A.C., Donev, A.N. (1992). Optimum Experimental Design. Oxford: Clarendon Press.Atkinson, A.C., Fedorov, V.V. (1988). “The optimum design of experiments in the presence of

uncontrolled variability and prior information.” In Optimal Design and Analysis of Experiments.Dodge, Y., Fedorov, V.V., Wynn, H. (editors). Amsterdam: North-Holland. 327–344.

Beal, S.L., Sheiner, L.B. (1992). NONMEM User’s Guide. NONMEM Project Group. SanFrancisco: University of California.

Bezeau, M., Endrenyi, L. (1986). “Design of experiments for the precise estimation of dose-responseparameters: the Hill equation.” Journal of Theoretical Biology. 123, 415–430.

Box, G.E.P., Hunter, W.G. (1965). “Sequential design of experiments for nonlinear models.”Proceedings of IBM Scientific Computing Symposium. Korth, J.J. (editor). White Plains, NewYork: IBM. 113–137.

Chaloner, K., Verdinelli, I. (1995). “Bayesian experimental design: a review.” Statistical Science. 10,273–304.

Chernoff, H. (1953). “Locally optimal designs for estimating parameters.” The Annals ofMathematical Statistics. 24, 586–602.

Cox, D.R. (1970). The Analysis of Binary Data. London: Chapman and Hall.

Page 207: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

194 Pharmaceutical Statistics Using SAS: A Practical Guide

Cramer, J.S. (1991). The LOGIT Model: An Introduction for Economists. London: Edward Arnold.D’Argenio, D.Z., Schumitzky, A. (1997). ADAPT II Users Guide:

Pharmacokinetic/Pharmacodynamic Systems Analysis Software. Biomedical SimulationsResource. Los Angeles: University of Southern California.

Downing, D.J., Fedorov, V.V., Leonov, S.L. (2001). “Extracting information from the variancefunction: optimal design.” In MODA6–Advances in Model-Oriented Design and Analysis.Atkinson, A.C., Hackl, P., Muller, W.G. (editors). Heidelberg: Physica-Verlag. 45–52.

Dragalin, V., Fedorov V. (2006). “Adaptive designs for dose-finding based on efficacy-toxicityresponse,” Journal of Statistical Planning and Inference. 136, 1800–1823.

Dragalin, V., Fedorov, V.V., Wu, Y. (2006). “Optimal design for bivariate probit model.” GSK BDSTechnical Report 2006-01. Collegeville, PA: GlaxoSmithKline.http://www.biometrics.com/downloads/TR 2006 01.pdf

Fan, S.K, Chaloner, K. (2001). “Optimal design for a continuation-ratio model.” InMODA6–Advances in Model-Oriented Design and Analysis. Atkinson, A.C., Hackl, P., Muller,W.G. (editors). Heidelberg: Physica-Verlag. 77–85.

Fedorov, V.V. (1972). Theory of Optimal Experiment. New York: Academic Press.Fedorov, V.V., Gagnon, R.C., Leonov, S.L. (2002). “Design of experiments with unknown

parameters in variance.” Applied Stochastic Models in Business and Industry. 18, 207–218.Fedorov, V.V., Hackl, P. (1997). Model-Oriented Design of Experiments. New York: Springer-Verlag.Fedorov, V.V., Leonov, S.L. (2001). “Optimal design of dose response experiments: a

model-oriented approach.” Drug Information Journal. 35, 1373–1383.Fedorov, V., Leonov, S., (2005). “Response-driven designs in drug development.” In Applied

Optimal Designs. Wong, W.K., Berger, M. (editors). Chichester: Wiley. 103–136.Finney, D.J. (1971). Probit Analysis. Third Edition. Cambridge: Cambridge University Press.Finney, D.J. (1976). “Radioligand assay.” Biometrics. 32, 721–740.Ford, I., Torsney, B., Wu, C.F.J. (1992). “The use of a canonical form in the construction of locally

optimal designs for non-linear problems.” Journal of the Royal Statistical Society. Series B. 54,569–583.

Gagnon, R., Leonov, S. (2005). “Optimal population designs for PK models with serial sampling.”Journal of Biopharmaceutical Statistics. 15, 143–163.

Gibaldi, M., Perrier, D. (1982). Pharmacokinetics. Second edition. New York: Marcel Dekker.Hedayat, A.S., Yan, B., Pezutto, J.M. (1997). “Modeling and identifying optimum designs for fitting

dose-response curves based on raw optical density data.” Journal of American StatisticalAssociation. 92, 1132–1140.

Heise, M.A., Myers, R.H. (1996). “Optimal designs for bivariate logistic regression.” Biometrics. 52,613–624.

Hill, P.D.H. (1980). “D-optimal designs for partially nonlinear regression models.” Technometrics.22, 275–276.

Kallen, A., Larsson, P. (1999). “Dose response studies: how do we make them conclusive?” Statisticsin Medicine. 18, 629–641.

Karpinski, K.F. (1990). “Optimality assessment in the enzyme-linked immunosorbent assay(ELISA).” Biometrics. 46, 381–390.

Kiefer, J., Wolfowitz, J. (1960). “The equivalence of two extremum problems.” Canadian Journal ofMathematics. 12, 363–366.

Kotz, S., Balakrishnan, N., Johnson, N.L. (2000). Continuous Multivariate Distributions. SecondEdition. Volume 1. New York: Wiley.

Lesaffre, E., Molenberghs, G. (1991). “Multivariate probit analysis: A neglected procedure inmedical statistics.” Statistics in Medicine. 10, 1391–1403.

Magnus, J.R., Neudecker, H. (1988). Matrix Differential Calculus with Applications in Statistics andEconometrics. New York: Wiley.

Page 208: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 7 Optimal Design of Experiments in Pharmaceutical Applications 195

McCullagh, P., Nelder, J. (1989). Generalized Linear Models. Second edition. New York: Chapmanand Hall.

Mentre, F., Mallet, A., Baccar, D. (1997). “Optimal design in random-effects regression models.”Biometrika. 84, 429–442.

Muirhead, R. (1982). Aspects of Multivariate Statistical Theory. New York: Wiley.Ogungbenro, K., Graham, G., Gueorguieva, I., Aarons, L. (2005). “The use of a modified Fedorov

exchange algorithm to optimise sampling times for population pharmacokinetic experiments.”Computer Methods and Programs in Biomedicine. 80, 115–125.

Pukelsheim, F. (1993). Optimal Design of Experiments. New York: Wiley.Rao, C.R. (1973). Linear Statistical Inference and Its Applications. Second edition. New York:

Wiley.Retout, S., Mentre, F. (2003). “Optimization of individual and population designs using S-Plus.”

Journal of Pharmacokinetics and Pharmacodynamics. 30, 417–443.Silvey, S.D. (1980). Optimal Design. London: Chapman and Hall.Sitter, R.R., Wu, C.F.J. (1993). “Optimal designs for binary response experiments: Fieller, D, and

A criteria.” Scandinavian Journal of Statistics. 20, 329–341.Torsney, B., Musrati, A.K. (1993). “On the construction of optimal designs with applications to

binary response and to weighted regression models.” In Model-Oriented Data Analysis. Muller,W.G., Wynn, H.P., Zhigljavsky, A.A. (editors). Heidelberg: Physica-Verlag. 37–52.

White, L.V. (1973). “An extension of the general equivalence theorem to nonlinear models.”Biometrika. 60, 345–348.

White, L.V. (1975). The optimal design of experiments for estimation in nonlinear model. Ph. D.thesis, University of London.

Wu, C.F.J. (1988). “Optimal design for percentile estimation of a quantal-response curve.” InOptimal Design and Analysis of Experiments. Dodge, Y., Fedorov, V.V., Wynn, H.P. (editors).North Holland: Elsevier. 213–223.

Wu, Y., Fedorov, V.V., Propert, K. J. (2005). “Optimal design for dose response using betadistributed responses.” Journal of Biopharmaceutical Statistics. 15, 753–771.

Wynn, H.P. (1970). “The sequential generation of D-optimal experimental designs.” Annals ofMathematical Statistics. 41, 1055–1064.

Zacks, S. (1996). “Adaptive designs for parametric models.” In Design and Analysis of Experiments.Handbook of Statistics. Volume 13. Ghosh, S., Rao, C.R. (editors). Amsterdam: Elsevier.151–180.

Page 209: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 210: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8

Analysis of HumanPharmacokinetic Data

Scott PattersonBrian Smith

8.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197

8.2 Bioequivalence Testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199

8.3 Assessing Dose Linearity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204

8.4 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209

This chapter introduces pharmacokinetic terminology and also describes the collection,measurement, and statistical assessment of pharmacokinetic data obtained in Phase I andclinical pharmacology studies. It focuses on the following two topics that play a key role inpharmacokinetic analysis: testing for bioequivalence and assessing dose linearity. Thechapter establishes a basic framework, and the examples demonstrate how to use restrictedmaximum likelihood models in the MIXED procedure to examine the most commonpharmacokinetic data of practical interest.

8.1 IntroductionWhen a tablet of drug is taken orally, in general, it reaches the stomach and begins todisintegrate and is absorbed (A). When dissolved into solution in the stomach acid, thedrug is passed on to the small intestine (Rowland and Tozer, 1980). At this point, some ofthe drug will pass through and be eliminated (E) from the body. Some will be metabolized(M) into a different substance in the intestine, and some drug will be picked up by thewalls of the intestine and distributed (D) into the body. This last bit of drug substancepasses through the liver first, where it is also often metabolized (M). The drug substancethat remains then passes through the liver and reaches the bloodstream, where it iscirculated throughout the body.

Pharmacokinetics (PK) is the study of absorption, distribution, metabolism, andexcretion (ADME) properties (Atkinson et al., 2001). Following oral administration, thedrug is held to undergo these four stages prior to being completely eliminated from thebody. PK is also the study of what the body does to a drug, (as opposed to what a drugdoes to the body).

Measurement is central to PK, and the most common method is to measure by means ofblood sampling how much drug substance has been put into the body (i.e., dose) relative to

Scott Patterson is Director, Statistical Sciences, GlaxoSmithKline Pharmaceuticals, USA. Brian Smith is Principal Bio-statistician II, Medical Sciences Biostatistics, Amgen, USA.

Page 211: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

198 Pharmaceutical Statistics Using SAS: A Practical Guide

how much drug reached the systemic circulation. For bioequivalence trials (U.S. Food andDrug Administration (FDA), 2003), at least 12 samples are collected over time followingdosing with an additional sample collected prior to dosing.

As the drug is absorbed and distributed, the plasma concentration rises and reaches amaximum (called the Cmax or maximum concentration). Plasma levels then decline untilthe body completely eliminates the drug from the body. The overall exposure to drug ismeasured by computing the area under the plasma concentration curve (AUC). AUC isderived in general by computing the area under each time interval and adding up all theareas. Other common summary measures are:

• Tmax (time of maximum concentration),• T1/2 (half-life of drug substance)

More details of techniques used in the derivation of AUC may be found in Yeh and Kwan(1978). Cmax and Tmax are derived by inspection of the data, and T1/2 is estimated by linearregression in accordance with the description of Rowland and Tozer (1980).

In this chapter, we will concentrate on the modeling of AUC and Cmax using PROCMIXED (note that, in general, the methods described are applicable to other summarymeasures). It is acknowledged that AUC and Cmax are log-normally distributed (Crow andShimizu, 1988) for the purposes of this discussion in accordance with the findings ofWestlake (1986), Lacey (1995), Lacey et al. (1997), and Julious and Debarnot (2000). ThusAUC and Cmax data are analyzed under ln (natural-logarithmic) transformation (Box andCox, 1964).

Analysis of ln-transformed AUC and Cmax data in clinical pharmacology follows thegeneral principles of analysis of repeated-measures, cross-over data using mixed effect linearmodels (Jones and Kenward, 2003; Wellek, 2003; Senn, 2002; Chow and Liu, 2000; Voneshand Chinchilli, 1997; Milliken and Johnson, 1992). To illustrate this assumption, consider astudy with n subjects and p periods where an observation y (either AUC or Cmax, etc.) isobserved for each subject in each period. Y may be expressed as a pn-dimensional responsevector. Then, in matrix notation, this model can be expressed as

Y = Xβ + Z1u + Z2e,

where Xβ is the usual design and fixed effects matrix, u is multivariate normal withexpectation 0 and variance-covariance matrix Ω, i.e.,

u ∼ MV N(0,Ω),

and

e ∼ MV N(0,Λ),

and u is independent of e. Subjects are assumed to be independent, and Z1 and Z2 aredesign matrices used to construct the variance-covariance structure as appropriate to thestudy design and desired variance components. The RANDOM and REPEATEDstatements in PROC MIXED are included as appropriate to calculate the desiredvariance-covariance structure.

Restricted maximum likelihood modeling (REML, Patterson, 1950; Patterson andThompson, 1971) is applied to calculate unbiased variance estimates and the degrees offreedom (Kenward and Roger, 1997) are used to calculate appropriate degrees of freedomfor any estimates or contrasts of interest.

Nonparametric statistical analysis of cross-over data will not be discussed further in thischapter but can be accomplished using SAS. Readers interested in such techniques shouldsee Jones and Kenward (2003), Wellek (2003), Chow and Liu (2000) and Hauschke et al.(1990) for more details.

Page 212: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 199

Other topics of general interest to readers of this chapter include PopulationPharmacokinetics (FDA Guidance, 1999) and Exposure-Response Modeling (FDA Guidance,2003). SAS is not generally used to support such nonlinear mixed-effect analyses, but isused in data management support (which will not be discussed here). It is theoreticallypossible to do such nonlinear mixed-effects modeling using the NLMIXED procedure, andwe refer readers interested in such analyses to Atkinson et al. (2001), Sheiner et al. (1989),Sheiner (1997), Sheiner and Steimer (2000), and Machado et al. (1999) for moreinformation on these topics.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html

8.2 Bioequivalence TestingBioequivalence testing is performed to provide evidence that a new formulation of drugsubstance (e.g., a new tablet) is equivalent in vivo to an existing formulation of drugproduct. Such testing is used by manufacturers of new chemical entities when makingchanges to a formulation of drug product used in confirmatory clinical trials, andadditionally is used by the generic pharmaceutical industry to secure market access atpatent expiration of an existing marketed product.

The new formulation (T) is generally studied relative to the existing formulation (R) ina randomized, open-label, cross-over trial (Jones and Kenward, 2003; Wellek, 2003; Senn,2002; Chow and Liu, 2000). The structure of testing the question of bioequivalence is:

H01 : μT − μR ≤ − ln 1.25,

H02 : μT − μR ≥ ln 1.25,

where μT is the adjusted mean for the test formulation and μR is the adjusted mean for thereference formulation on the natural log scale. The factor ln 1.25 was chosen by regulatoryagencies (FDA, 1992; Barrett et al., 2000) and each one-sided test is performed at a 5%significance level without adjustment for multiplicity (Hauck et al., 1995).

Both tests must reject the null hypotheses for AUC and Cmax in order for bioequivalenceto be declared. This testing procedure is referred to as the TOST (two-one sided testing, seeSchuirmann, 1987) assessment of average bioequivalence and constitutes the current FDAstandard approach. See the FDA guidance page, http://www.fda.gov/cder/guidance/ formore details. In practice, a 90% confidence interval for μT − μR is derived for theln-transformed AUC and Cmax, using a model appropriate to the study design. If bothintervals fall completely within the interval (− ln 1.25, ln 1.25), bioequivalence is concluded.This procedure is designated the average bioequivalence (ABE) because only the test andreference means are compared. A summary of other approaches and issues of interest inbioequivalence testing may be found in Hauck and Anderson (1992), Anderson and Hauck(1996), Benet (1999) and Zariffa and Patterson (2001). This type of approach is termed aconfirmatory bioequivalence assessment for the purposes of this chapter.

This type of approach to data analysis is applied to clinical PK data in a variety ofother setting in exploratory clinical pharmacology research. Examples include drug-druginteraction trials, food effect assessment and comparison of rate and extent ofbioavailability in renal-impaired and hepatic insufficient populations. See the FDAguidance page, http://www.fda.gov/cder/guidance/ for details. In these studies, therange of plausible values as expressed by a confidence interval is used to assess the degreeof equivalence or comparability, depending upon the setting. A TOST need not necessarilybe performed. Under such an approach, confidence level (Type I error rate) is termedconsumer or regulator risk, i.e., the risk of the regulator agency in making an incorrectdecision. Though often a pre-specified equivalence limit is difficult or impossible to define

Page 213: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

200 Pharmaceutical Statistics Using SAS: A Practical Guide

prior to study initiation, thus inhibiting the ability of study sponsors to adequately ensureadequate power to demonstrate equivalence if such is desired, power is of less concern whenassessing the results of such studies than the confidence level. This approach to statisticalinference gives regulator agencies an easy standard under which to assess the results ofsuch exploratory pharmacokinetic studies.

We now turn to an example of a confirmatory bioequivalence trial.

EXAMPLE: Two-period cross-over study with test and reference formulationsConsider the following 2 × 2 cross-over study where a test formulation was compared to areference formulation in approximately 50 normal healthy volunteer subjects.

The AUC data set, reproduced with permission from Patterson (2001), summarizesAUC values derived for the test and reference periods (the data set can be found on thebook’s companion Web site). The SUBJECT variable is the subject’s ID number, theSEQUENCE variable codes the treatment sequence (RT denotes “‘reference-test” and TRdenotes “test-reference”) and, lastly, the AUCT and AUCR variables contain AUC valueswhen given Test and Reference formulations, respectively, in [ng/mL/h].

Program 8.1 uses PROC MIXED to fit a random-intercept model (Jones and Kenward,2003) to the AUC data set once the AUC data are formatted by subject, sequence, period,and formulation (program not shown).

Program 8.1 Comparison of AUC values in the two-period cross-over study with test and referenceformulations

data lnauc(keep=subject sequence period formula lnauc);set auc;lnauc=log(auc);

proc mixed data=lnauc method=reml;class sequence subject period formula;model lnauc=sequence period formula/ddfm=kenwardroger;random subject(sequence);estimate ’T-R’ formula -1 1/cl alpha=0.10;run;

Output from Program 8.1

Cov Parm Estimate

subject(sequence) 1.5921Residual 0.1991

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

sequence 1 44.8 0.12 0.7254period 1 43.1 0.37 0.5463formula 1 43.1 0.92 0.3424

Page 214: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 201

Estimates

StandardLabel Estimate Error DF t Value Pr > |t|

T-R 0.09023 0.09399 43.1 0.96 0.3424

Label Alpha Lower Upper

T-R 0.1 -0.06776 0.2482

Output 8.1 displays the estimated difference between the test and reference formulationsin the ln-transformed AUC with a 90% confidence interval. The 90% confidence interval is(−0.0678, 0.2482). As insufficient information is present to reject H02, bioequivalence wasnot demonstrated. No significant differences in sequence (p = 0.7254), period (p = 0.5463),and formulation (p = 0.3424) were detected. Between-subject variance in ln-AUC was1.5921, and within-subject variance was 0.1991.

Alternatively we can use the following RANDOM statement to explicitly specify arandom-intercept model:

random int/subject=subject(sequence);

Specifying the RANDOM statement results in equivalent findings to the above.These are the models required by FDA (1992) for the assessment of bioequivalence using

a 2 × 2 cross-over design. Note that the Huyhn-Feldt condition (Hinkelmann andKempthorne, 1994) is applied to the variance components using this model.

We may also use the REPEATED statement in PROC MIXED if the total variance(between plus within subject variation) of test and reference formulations and theircovariance are of interest in a 2 × 2 cross-over study:

proc mixed data=lnauc method=reml;class sequence subject period formula;model lnauc=sequence period formula/ddfm=kenwardroger;repeated formula/type=un subject=subject(sequence);estimate ’T-R’ formula -1 1/cl alpha=0.10;run;

Results of this last statement may differ slightly from the random-intercept model abovewhen data are missing. It is unlikely, but not impossible, that missing data can affectinference.

8.2.1 Alternative Designs in Bioequivalence TestingThe FDA guidance also allows for alternative designs to demonstrate bioequivalence. Onesuch design is the replicate cross-over design where each subject receives each formulationtwice, with each administration being separated by a wash-out period of five half-lives.

EXAMPLE: A replicate cross-over studyConsider the following replicate design cross-over study where test and referenceformulations were adminstered in a randomized, four-period cross-over design inapproximately 36 normal healthy volunteer subjects.

The RAUC data summarizes AUC values collected in the test and reference periods (thedata set can be found on the book’s companion Web site). The SUBJECT variable is thesubject’s ID number, SEQUENCE variable codes the treatment sequence (ABBA denotes

Page 215: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

202 Pharmaceutical Statistics Using SAS: A Practical Guide

“test-reference-reference-test” and BAAB denotes “reference-test-test-reference”) and,lastly, AUC1TEST, AUC2TEST, AUC1REF, AUC2REF variables contain AUC valueswhen given Test and Reference formulations in the first and second administrations,respectively, in [ng/mL/h].

The FDA guidance requires that the construction of the confidence interval for μT − μR

be appropriate to the study design. In the case of a replicate design model, thisrecommendation results in a requirement to specify a factor-analytic variance-covariancestructure for the between-subject variance components and calculation of within-subjectvariances for each formulation; see Patterson and Jones (2002) for more details.

Program 8.2 uses PROC MIXED to fit a factor-analytic variance-covariance structureusing the FA0(2) option to the ln-transformed AUC data by subject, sequence, period, andformulation. See FDA Guidance (2001) for a description of why this option is used.

Program 8.2 Analysis of AUC data from a replicate cross-over study with test and reference formulations

data lnauc(keep=subject sequence period formula lnauc);set rauc;lnauc=log(auc);

proc mixed data=lnauc method=reml;class sequence subject period formula;model lnauc=sequence period formula/ddfm=kenwardroger;random formula/type=FA0(2) subject=subject;repeated/group=formula subject=subject;estimate ’T-R’ formula 1 -1/cl alpha=0.1;ods output estimates=test;

data test;set test;lowerb=exp(lower); * Lower bound on original scale;upperb=exp(upper); * Upper bound on original scale;

proc print data=test noobs;var lowerb upperb;run;

Output from Program 8.2

Covariance Parameter Estimates

Cov Parm Subject Group Estimate

FA(1,1) subject 0.5540FA(2,1) subject 0.5542FA(2,2) subject 1.24E-17Residual subject formula A 0.09851Residual subject formula B 0.1110

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

sequence 1 35.6 0.09 0.7706period 3 106 2.28 0.0835formula 1 106 7.68 0.0066

Page 216: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 203

Estimates

StandardLabel Estimate Error DF t Value Pr > |t|

T-R -0.1449 0.05227 106 -2.77 0.0066

Label Alpha Lower Upper

T-R 0.1 -0.2316 -0.05816

lowerb upperb

0.79324 0.94350

Output 8.2 lists the findings. For ln-transformed AUC, a confidence interval of(−0.2316,−0.0582) is found. As insufficient information is present to reject H01 in this case,bioequivalence was not demonstrated. These limits are saved in the TEST data set using anODS statement and may be exponentiated in an additional DATA step if findings on theoriginal scale are desired. In this case, the lower and upper bounds are 0.7932 and 0.9425.

Within-subject variation for the test and reference formulations were 0.09851 and0.1110, respectively. Variation associated with subject-by-formulation was negligible, withan estimated standard deviation of 1.24E-17.

No significant evidence of sequence (p = 0.7706) or period (p = 0.0835) effects wasdetected; however, the formulations were observed to be significantly different (p = 0.0066).

The FDA guidance (2001) states that “In the Random statement, Type=FA0(2) couldpossibly be replaced by Type=CSH. This guidance recommends that Type=UN not beused, as it could result in an invalid (i.e., non-negative definite) estimated covariancematrix.”

There is however a similar issue with use of TYPE=FA0(2) or CSH. These areconstrained structures in that their application does not allow all positive definitecovariance structures to be estimable (Patterson and Jones, 2002). For example, in theCSH structure, the estimate for correlation, ρ, is constrained by PROC MIXED so that itlies in the interval −1 ≤ ρ ≤ 1. The FA0(2) structure similarly imposes constraints suchthat the estimate for the subject-by-formulation standard deviation is zero or greater.

The choice of FA0(2) is somewhat arbitrary and reflective in regulatory application ofGeorge Box’s statement “All models are wrong, but some are useful.” In applications, it hasbeen shown that this procedure protects the Type I error rate (of key concern to regulators)when variance estimates of interest are constrained to be positive or null (Patterson andJones, 2004). It has the additional benefit of providing variance-covariance estimates thatare readily interpretable (i.e., non-negative); however, statisticians using these proceduresshould recognize that the variance-covariance estimates may be biased as a consequence ofthis choice. Note that restricted maximum likelihood modeling (REML) is also specified asa recommended option, and those desiring to apply a different method should first talkwith the FDA. Although REML estimates do possess less bias than maximum likelihoodestimates, they do not produce estimates which maximize the likelihood function. TheFDA has thus chosen a model which is readily interpretable and potentially biased, butwhich protects their risk of making a false positive decision of bioequivalence.

Note that if the TYPE=UN option is specified in Program 8.2, removing this constraint,a confidence interval of (−0.2220,−0.0710) is derived as subject-by-formulation variation isestimated to be negative.

Page 217: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

204 Pharmaceutical Statistics Using SAS: A Practical Guide

8.3 Assessing Dose LinearityAs discussed in the introduction, when an oral tablet is taken it will be absorbed,distributed, metabolized and eliminated. Imagine that the body is made up of manydifferent compartments. The drug amount that enters circulation is then bothsimultaneously distributed into and eliminated from these compartments. Typically, a drugis eliminated from the body through urine or feces. In order to facilitate this process, theliver often metabolizes the compound to a chemical form that is more readily eliminated. Ifall of the eliminations from one compartment to another are proportional to theconcentration of the compound in the compartment, the drug has dose linearity. Doselinearity implies that the elimination rate from the body is proportional as well.

The following two conditions are therefore implied. First, the elimination rate isproportional to drug concentration with k denoting the proportion. This constant is oftenreplaced with clearance. As can be seen, dose linearity implies that clearance is a constant.Second, with repeated dosing there exists a point in time in which there is an equilibriumbetween the amount of drug that enters the body and the amount that leaves. Once thisequilibrium has been reached, the drug is said to have achieved steady state.

As can be seen, it would be impossible to really tell if a drug were truly dose linear;however, a drug that has dose linearity (or that at least approximates dose linearity)should have certain properties. One of these is a proportional increase in concentration inthe blood or plasma as measured with AUC and Cmax, with increasing dose. This situationis often referred to as dose proportionality. A second property is that the clearance ought tobe stationary with time. Both of these properties can be studied in human pharmacokineticstudies.

8.3.1 Assessing Dose ProportionalityDose proportionality is a proportional increase in concentration as measured with AUCand Cmax, with increasing dose. Using AUC, this implies that

AUC = αd,

where d represents the dose. There are many different models to examine doseproportionality. A discussion to the relative merits and demerits of these models can befound is Smith (2004). This section will focus on the following model

lnAUC = α + β ln d.

This model is referred to as the power model on an ln-transformed AUC. The same model isused for Cmax as discussed in the previous section. Notice that when we exponentiate bothsides of this equation we have

AUC = exp(α)dβ.

Thus, when β = 1, the drug is dose proportional.In assessing dose proportionality, we may first consider a hypothesis test of the following

H0 : β = 1.

We may consider declaring dose proportionality if we fail to reject this hypothesis. Smithet al. (2000) argue that it is much more natural to think of dose proportionality as anequivalence problem, implying that the structure for testing dose proportionality should be

H01 : β ≤ 1 − t,

H02 : β ≥ 1 + t.

Unlike bioequivalence, the equivalence region currently has no set regulatory standard.Smith et al. (2000) considers the following:

Page 218: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 205

1. Examine dose-normalized concentrations.2. Define some high dose (h) and low dose (l), where r = h/l, for which we wish to

examine dose proportionality.3. Set the following structure for testing the equivalence of the dose-normalized

concentrations:

H03 : μh − μl ≤ − ln θ,

H04 : μh − μl ≥ ln θ,

where μh and μl denote the dose-normalized mean of the high dose and low doses on thenatural log scale, respectively.

Smith et al. (2000) show that this structure is equivalent to the structure of H01 and H02with t = ln θ/ ln r. Notice that t → ∞ when r → 1 and t → 0 when r → ∞. Thus, thereexists a set of r’s such that both H01 and H02 will be rejected.

The largest of these r’s becomes the largest ratio of doses for which dose proportionalitycan be declared. Smith et al. (2000) show that if H01 and H02 are tested at the α level andthat if L and H define the lower and upper limit of a (1 − 2α)100% confidence interval, thislargest ratio ρ1 is

ρ1 = θ(1/max(1−L,U−1)).

Furthermore, Smith et al. (2000) describe three possible conclusions that can be drawnabout dose proportionality:

1. Definitely dose proportional.2. Definitely not dose proportional.3. Inconclusive.

“Definitely not dose proportional” is concluded if either the null hypothesis H01 or thenull hypothesis H02 cannot be rejected. Smith et al. (2000) show that sometimes there existsa smallest ratio, ρ2, in which dose proportionality definitely does not exist and is given by

ρ2 = θ(1/ max(L−1,1−U)).

It should be pointed out that this equation cannot be solved unless L > 1 or U < 1.Data for examination of dose proportionality most often comes from first human, dose

escalation type trials in which randomization-to-period is not done. Occasionally, astand-alone study is performed to ensure that period effects do not confound inference.

EXAMPLE: First human-dose studyConsider the simulated data from a hypothetical first human dose trial (FHDAUC data setcan be found on the book’s companion Web site). Here 16 subjects received ascendingdoses of drug, and AUC(0-∞) [ng/mL/h] was measured after each administration.

Program 8.3 shows how to perform the dose proportionality analysis of the AUC datausing a random-intercept model in PROC MIXED.

Page 219: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

206 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 8.3 Analysis of AUC data from the first human-dose study

data fhdauc;set fhdauc;lndose=log(dose);lnauc=log(auc);study=1;

proc mixed data=fhdauc method=reml;class subject;model lnauc=lndose/s ddfm=kenwardroger cl alpha=0.1;random subject;ods output solutionf=one;

data one;set one;if effect=’lndose’ then delete;rho1=(4/3)**(1/max(1-lower,upper-1));if lower < 1 and upper > 1 then rho2=.;else rho2=(4/3)**(1/max(lower-1,1-upper));

proc print data=one noobs;var rho1 rho2;run;

Output from Program 8.3

Covariance Parameter Estimates

Cov Parm Estimate

subject 0.04175Residual 0.01861

Solution for Fixed Effects

StandardEffect Estimate Error DF t Value Pr > |t|

Intercept -0.2583 0.07518 36.9 -3.44 0.0015lndose 1.0395 0.01235 30.7 84.20 <.0001

Effect Alpha Lower Upper

Intercept 0.1 -0.3851 -0.1314lndose 0.1 1.0186 1.0605

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

lndose 1 30.7 7088.96 <.0001

rho1 rho2

116.630 5344869.90

Page 220: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 207

Output 8.3 shows that β significantly differs from unity (p < 0.0001) with 90%confidence bounds of (1.0186, 1.0605). Between-subject variation in ln-AUC was 0.0418,and within-subject variation was observed to be 0.0186.

Output 8.3 also extracts information from PROC MIXED in order to calculate ρ1 andρ2. Notice that θ = 4/3 in Program 8.3. The choice for θ is user-defined. In this case theresulting equivalence region is (0.75, 1.33), as compared to the equivalence region forbioequivalence of (0.80, 1.25). Dose proportionality is concluded as the 90% confidenceinterval for β falls within these limits.

In this first human-dose study, one derives ρ1 = 117 and ρ2 = 5, 340, 000. One wouldinterpret this to mean that dose proportionality has been demonstrated up to a 117-foldrange of doses. Said another way, if we knew the concentration (c) for some dose a, then agood prediction of the concentration for a dose 117a would be 117c. The value of ρ2 in thiscase is so large that it has little practical meaning. All and all, since it is unlikely in clinicalpractice to have available a 117-fold range of doses, this molecule demonstrates asignificant degree of dose proportionality.

Note that, in this example, ρ1 = 117 and the dose range studied is 200-fold. It is quitepossible for ρ1 to be greater than the dose range studied. This brings up the question ofhow we should interpret ρ1 if this is the case. Assume that in this example ρ1 turned out tobe 500. There would obviously be danger in saying that a good prediction of theconcentration for a dose 500a would be 500c, since we are obviously extrapolating beyondavailable data. With this said, however, it is still useful to think of ρ1 as a measure of doseproportionality. That is, a value of 500 indicates a greater degree of dose proportionalitythan a value of 300 would have.

There are certainly other covariance structures that could be chosen in Program 8.3. Forinstance, a random intercept and slope model can be generated with the followingRANDOM statement:

random subject subject*lndose;

8.3.2 Assessing Time Stationarity of ClearanceIn the beginning of this section, clearance was defined as the rate of elimination divided bythe concentration. When an oral dose is administered, only a fraction of the total dose isabsorbed by the body. It can be shown that for a single dose of a drug that

Clearance =Fd

AUC(0 − ∞),

where F is the fraction adsorbed, d is the dose and AUC(0 − ∞) is what has been calledAUC so far in this chapter. On the other hand, when a drug is at steady state, it can beshown that

Clearance =Fd

AUC(0 − τ),

where τ is the dosing interval (24 hours if dosed once a day), d is the amount of drugdelivered at each dosing interval, and AUC(0 − τ) is the area under the concentration whenthe drug is at steady state. The implication of these two formulas is that ifAUC(0 − ∞) = AUC(0 − τ) for one dose, then clearance does not change with repeateddosing. If this is true for all doses, then clearance is said to be stationary. Dose linearityimplies that clearance is a constant, which implies stationarity.

If the half-life of a drug is short enough, it may be possible to calculate AUC(0 − ∞) foreach individual in the first day of dosing of a multiple dose study and find AUC(0-τ) at thelast day. Subjects must be at steady state and measures should be taken to ensure this.

Page 221: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

208 Pharmaceutical Statistics Using SAS: A Practical Guide

The shorter the half-life, the quicker steady state can be achieved. Thus, since we need ashort half-life to be able to do this, the drug would most likely be at steady state. Having ashort enough half-life to be able to use day 1 data, however, is pretty unlikely.

Another possibility is doing the first human dose study in the same subjects as themultiple dose study. Usually, when this is done, these studies are combined into oneprotocol. Again, however, the opportunity to do this does not present itself very often. Thereason these options are appealing is it allows us to use a subject as its own control. Themost common case, however, is that the first human dose study and multiple dose studyare done in separate groups of subjects. We can, however, still combine this information toexamine stationarity of clearance.

EXAMPLE: Multiple-dose studyConsider the MDAUC data set that can be found on the book’s companion Web site. Thedata set contains simulated data from a hypothetical multiple-dose study. This studytested the compound examined in the first human-dose trial described above. It is assumedthat the multiple-dose study was performed in a different set of subjects from the firststudy. Here, 16 subjects were dosed with 20 to 200 mg of drug repeatedly for several days,and AUC(0-τ) [ng/mL/h] was measured over the dosing interval on the final day.

Program 8.4 examines stationarity of clearance in the multiple-dose study using PROCMIXED. Stationarity of clearance would be concluded if the estimated geometric meansfrom the 20 mg first human dose trial and estimated geometric means from the 20 mgmultiple dose trial are equivalent and the estimated geometric means from the 200 mg firsthuman dose trial and estimated geometric means from the 200 mg multiple dose trial areequivalent.

The PROC MIXED code in Program 8.4 fits a separate power model for both studies.The ESTIMATE statements are then used to derive estimates for the single dose meanAUC at 20 mg, the multiple dose mean AUC at 20 mg, their ratio, the single dose mean at200 mg, the multiple dose mean at 200 mg, and their ratio, respectively. These estimatesare saved in the OUT data set and then exponentially transformed to the original scale inorder to compute 90% confidence intervals for each scenario. Program 8.4 suppresses thestandard PROC MIXED output using ODS LISTING statements. Note that the log-dosesof 20 and 200 mg are denoted as 2.995732 and 5.298317, respectively.

Program 8.4 Analysis of AUC data from the multiple-dose study

data mdauc;set mdauc;lnauc=log(auc);lndose=log(dose);study=2;

* Merge the data from the first human-dose and multiple-dose studies;data cl;

set fhdauc mdauc;ods listing close;proc mixed data=cl method=reml;

class subject study;model lnauc=lndose study lndose*study/ddfm=kenwardroger cl alpha=0.1;random subject*study;estimate ’Single dose AUC 20 mg’ intercept 1 lndose 2.995732

study 1 lndose*study 2.995732;estimate ’Multiple dose AUC 20 mg’ intercept 1 lndose 2.995732

study 1 lndose*study 0 2.995732;estimate ’Ratio single dose:multiple dose 20 mg’ study 1 -1

lndose*study 2.995732 -2.995732/cl alpha=0.1;

Page 222: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 209

estimate ’Single dose AUC 200 mg’ intercept 1 lndose 5.298317study 1 lndose*study 5.298317;

estimate ’Multiple dose AUC 200 mg’ intercept 1 lndose 5.298317study 0 1 lndose*study 0 5.298317;

estimate ’Ratio single dose:multiple dose 200 mg’ study 1 -1lndose*study 5.298317 -5.298317/cl alpha=0.1;

ods output estimates=out;data out;

set out;gmean=exp(estimate);lbound=exp(lower);ubound=exp(upper);drop estimate stderr df tvalue alpha lower upper;

proc print data=out noobs;ods listing;run;

Output from Program 8.4

Label Probt gmean lbound ubound

Single dose AUC 20 mg <.0001 17.385 15.617 19.354Multiple dose AUC 20 mg <.0001 16.738 13.697 20.455Ratio single dose:multiple dose 20 mg 0.7371 1.039 0.860 1.255Single dose AUC 200 mg <.0001 190.365 170.987 211.939Multiple dose AUC 200 mg <.0001 214.759 177.887 259.273Ratio single dose:multiple dose 200 mg 0.2667 0.886 0.740 1.062

Output 8.4 lists the estimated geometric means and associated 90% confidence limits.The 90% confidence interval of the ratio of geometric means for 20 mg is (0.860, 1.255) andthe 90% confidence interval of the ratio of geometric mean for 200 mg is (0.740, 1.062).Although neither confidence interval meets the strict bioequivalence (0.8, 1.25) criteria, atthis point of drug development, given the limited amount of data, stationarity of clearancewould be assumed, until and unless compelling future data indicated otherwise.

Since dose proportionality can be concluded and dose stationarity seems highly tenable,this molecule seems to exhibit dose linearity.

8.4 SummaryIn this chapter, pharmacokinetic terminology frequently encountered in clinical trialassessment of human pharmacokinetics was reviewed, and the collection, measurement, andstatistical assessment of pharmacokinetic data obtained in Phase I and clinicalpharmacology studies were described. REML models were used to examine the mostcommon pharmacokinetic data of practical interest, and tests for assessing bioequivalenceand dose linearity were developed and implemented using PROC MIXED.

ReferencesAtkinson, A., Daniels, C., Dedrick, R., Grudzinskas, C., Markey, S. (2001). Principles of Clinical

Pharmacology. San Diego: Academic Press.Anderson, S., Hauck W.W. (1996). “The transitivity of bioequivalence testing: potential for drift.”

International Journal of Clinical Pharmacology and Therapeutics. 34, 369–374.Barrett, J.S., Batra, V., Chow, A., Cook, J., Gould, A.L., Heller, A., Lo, MW., Patterson, S.D.,

Smith, B.P., Stritar, J.A., Vega, J.M., Zariffa, N. (2000). “PhRMA Perspective on Population

Page 223: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

210 Pharmaceutical Statistics Using SAS: A Practical Guide

and Individual Bioequivalence and Update to the PhRMA Perspective on Population andIndividual Bioequivalence.” Journal of Clinical Pharmacology. 40, 561–572.

Benet, L.Z. (1999). “Understanding Bioequivalence Testing.” Transplantation Proceedings. 31,Supplement 1, 7S–9S.

Box, G.E., Cox, D.R. (1964). “An analysis of transformations.” Journal of the Royal StatisticalSociety, Series B. 26, 211–243.

Chow, S.C., Liu, J. (2000). Design and Analysis of Bioavailability and Bioequivalence Studies,second edition. New York: Marcel Dekker.

Crow, E.L., Shimizu, K. (1988). Lognormal distributions. New York: Marcel Dekker.FDA Guidance. (1992). Statistical procedures for bioequivalence studies using a standard two

treatment cross-over design.FDA Guidance. (February, 1999). Guidance for Industry: Population Pharmacokinetics.FDA Guidance. (January, 2001). Guidance for Industry: Statistical Approaches to Establishing

Bioequivalence.FDA Guidance. (April, 2003). Guidance for Industry: Exposure-Response Relationships—Study

Design, Data Analysis, and Regulatory Applications.Hauck, W.W., Anderson S. (1992). “Types of bioequivalence and related statistical considerations.”

International Journal of Clinical Pharmacology, Therapy, and Toxicology. 30, 181–187.Hauck, W.W., Hyslop, T., Anderson, S., Bois, F.Y., Tozer, T.N. (1995). “Statistical and regulatory

considerations for multiple measures in bioequivalence testing.” Clinical Research and RegulatoryAffairs. 12, 249–265.

Hauschke, D., Steinijans, V.W., Diletti, E. (1990). “A distribution free procedure for the statisticalanalysis of bioequivalence studies.” International Journal of Clinical Pharmacology, Therapy,and Toxicology. 28, 72–78.

Hinkelmann, K., Kempthorne, O. (1994). Design and Analysis of Experiments, Volume I:Introduction to Experimental Design. New York: Wiley.

Jones, B., Kenward M.G. (2003). Design and Analysis of Cross-over Trials, second edition. London:Chapman and Hall.

Julious, S., Debarnot, C. A-M. (1999). “Why are pharmacokinetic data summarized by arithmeticmeans?” Journal of Biopharmaceutical Statistics. 10, 55–71.

Kenward, M., Roger, J. (1997). “Small sample inference for fixed effects from restricted maximumlikelihood.” Biometrics. 33, 983–997.

Lacey, L.F., Keene, O.N., Bye, A. (1995). “Glaxo’s exprerience of different absorption rate metrics ofimmediate release and extended release dosage forms.” Drug Information Journal. 29, 821–840.

Lacey, L.F., Keene, O.N., Pritchard, J.F., Bye, A. (1997). “Common noncompartmentalpharmacokinetic variables: are they normally or log-normally distributed?” Journal ofBiopharmaceutical Statistics. 7, 171–178.

Machado, S., Miller, R., Hu, C. (1999). “A regulatory perspective on pharmacokinetic andpharmacodynamic modelling.” Statistical Methods in Medical Research. 8, 217–45.

Milliken, G.A., Johnson, D.E. (1992). Analysis of Messy Data, Volume 1: Designed Experiments,New York: Chapman and Hall.

Patterson, H.D. (1950). “The construction of balanced designs for experiments involving sequencesof treatments.” Biometrika. 39, 32.

Patterson, H.D., Thompson, R. (1971). “Recovery of Inter-block information when block sizes areunequal.” Biometrika. 58, 545–554.

Patterson, S.D. (2001). “A review of the development of biostatistical design and analysistechniques for assessing in vivo bioequivalence, Part 1.” Indian Journal of PharmaceuticalSciences. 63, 81–100.

Patterson, S.D., Jones, B. (2002). “Bioequivalence and the pharmaceutical industry.”Pharmaceutical Statistics. 1, 83–95.

Patterson, S.D., Jones, B. (2004). “Simulation assessments of statistical aspects of bioequivalence inthe pharmaceutical industry.” Pharmaceutical Statistics. 3, 13–23.

Page 224: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 8 Analysis of Human Pharmacokinetic Data 211

Rowland, M., Tozer, T.N. (1980). Clinical Pharmacokinetics Concepts and Applications.Philadelphia: Lea and Febiger.

Schuirmann, D.J. (1987). “A comparison of the two one sided tests procedure and the powerapproach for assessing the equivalence of average bioavailability.” Journal of Pharmacokineticsand Biopharmaceutics. 15, 657–680.

Senn, S. (2002) Cross-over Trials in Clinical Research, second edition. New York: John Wiley &Sons.

Sheiner L.B., Beal, S.L., Sambol, N.C. (1989). “Study Designs for Dose-Ranging.” ClinicalPharmacology and Therapeutics. 46, 63–77.

Sheiner, L.B. (1997). “Learning versus confirming in clinical drug development.” ClinicalPharmacology and Therapeutics. 61, 275–291.

Sheiner, L.B., Steimer, J-L. (2000). “Pharmacokinetic-pharmacodynamic modeling in drugdevelopment.” Annual Reviews of Pharmacology and Toxicology. 40, 67–95.

Smith, B., Vandenhende, F.R., DeSante, K., Farid, N., Welch, P., Callaghan, J., Forgue, S. (2000).“Confidence interval criteria for assessment of dose proportionality.” Pharmaceutical Research.17, 1278–1283.

Smith, B.P. (2004). “Assessment of Dose Proportionality.” Chapter 6, Part 2 of Pharmacokineticsin Drug Development: Clinical Study Design and Analysis, Volume 1. Bonate, P., Howard, D.,editors. Arlington,VA: AAPS Press. 363–382.

Vonesh, E.F., Chinchilli, V.M. (1997). Linear and Nonlinear models for the analysis of repeatedmeasurements. New York: Marcel Dekker.

Wellek, S. (2003) Testing Statistical Hypotheses of Equivalence. London: Chapman and Hall.Westlake, W.J. (1986). “Bioavailability and bioequivalence of pharmaceutical formulations.”

Biopharmaceutical Statistics for Drug Development. K. Peace, editor. New York: Marcel Dekker.329–352.

Yeh, K.C., Kwan, K.C. (1978). “A comparison of numerical integrating algorithms of trapezoidal,Legrange, and spline approximation.” Journal of Pharcokinetics and Biopharmaceutics. 6, 79–81.

Zariffa, N.M-D., Patterson, S.D. (2001). “Population and Individual Bioequivalence: Lessons fromReal Data and Simulation Studies.” Journal of Clinical Pharmacology. 41, 811–822.

Page 225: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 226: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9

Allocation in RandomizedClinical Trials

Olga KuznetsovaAnastasia Ivanova

9.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213

9.2 Permuted Block Randomization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214

9.3 Variations of Permuted Block Randomization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217

9.4 Allocations Balanced on Baseline Covariates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228

9.5 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233

This chapter discusses the operational aspects of randomization designs used in thepharmaceutical industry. Most of this chapter is devoted to the widely used permuted blockdesign and its variations. Allocation procedures that are balanced on baseline covariates,such as stratified randomization, and covariate-adaptive randomization, are also described.All methods are illustrated by examples that include SAS code to generate allocationsequences. Although all of the examples in this chapter refer to clinical trials, the describedrandomization methods can also be used in a non-clinical setting, i.e., in animal studies.

We refer the reader to Rosenberger and Lachin (2002) for a thorough coverage of otheraspects of randomization such as randomization-based inference and covariate-adjustedanalysis. A chapter in Senn’s book (1997) offers excellent insights on randomization inclinical trials.

9.1 IntroductionRandomization, an allocation of subjects to treatment regimens using a random element, isan essential component of clinical trials. Randomization promotes comparability of thetreatment groups with respect to known as well as unknown covariates and thus reducesthe chance for bias in the evaluation of the treatment effect. It can also serve as a basis fora randomization approach to inference (Rosenberger and Lachin, 2002).

Several types of bias are considered in the context of randomization. Selection biasoccurs in an unmasked (unblinded) trial when the investigator, either consciously orotherwise, uses knowledge of the upcoming treatment assignment to help decide whom toenroll (Blackwell and Hodges, 1957). Observer bias is the bias in the evaluation of responses

Olga Kuznetsova is Associate Director, Clinical Biostatistics, Merck, USA. Anastasia Ivanova is Associate Professor, De-partment of Biostatistics, University of North Carolina-Chapel Hill, USA.

Page 227: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

214 Pharmaceutical Statistics Using SAS: A Practical Guide

to a treatment that can occur if some of the assignments are known to the investigator.Imbalance in a covariate strongly associated with the study outcome can also bias theresults. Although it is possible to adjust for known covariates, imbalance in unobservedcovariates can lead to accidental bias (Efron, 1971). Various allocation procedures differ inhow susceptible they are to these types of bias, and that fact influences the choice of anallocation procedure for a clinical trial. Another consideration that needs to be taken intoaccount when selecting an allocation procedure is the procedure’s ability to achieve atargeted allocation ratio.

Complete randomization, in which each subject is allocated at random with theprobability determined by the targeted allocation ratio, is totally unpredictable and doesnot lead to selection bias in a trial unmasked to the investigator. Its significant drawback,though, is the risk of undesirable imbalance in the number of subjects allocated to eacharm. Such imbalance can negatively affect the power of treatment comparisons, especiallywhen the total number of subjects in the trial is small. Also, if the subjects’ prognosticfactors exhibit a time trend during the course of the trial, a considerable imbalance intreatment assignments throughout the trial can lead to accidental bias. Restrictedrandomization implemented through a permuted block design (Rosenberger and Lachin,2002) provides a good balance in the treatment assignments throughout the enrollment andis more commonly used. Other designs that maintain a good balance in treatmentassignments at any time and have low potential for selection bias have recently beensuggested (Berger et al., 2003).

After an appropriate randomization procedure is selected, the sequence of randomassignments for the study subjects is generated and documented in the randomization(allocation) schedule.

It might be desirable to maintain balance in important prognostic baseline covariates.Balance is typically achieved through stratified randomization, in which a separaterestricted allocation schedule is prepared for each combination of levels of the stratificationfactors. Alternatively, balance in baseline covariates can be maintained with a dynamicallocation, in which a new subject is assigned to the treatment group that results in thebest balance (in some sense) across his or her set of covariates. A fixed allocation schedulecannot be prepared for dynamic procedures, as the sequence of treatment assignmentsdepends on the covariates of the subjects entering the trial.

The randomization schedule or a sequence of treatment assignments for a dynamicallocation procedure can be easily generated with SAS software, e.g., using the PLANprocedure or random number generators. The flexibility of the PLAN procedure oftenallows the same schedule to be generated in many different ways; we will cover a variety ofoptions in the examples throughout this chapter.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

9.2 Permuted Block RandomizationPermuted block randomization is commonly used in clinical trials to allocate subjects to thetreatment arms in required ratios. It is well accepted in the clinical community, providesgood balance in treatment assignments, and supports the needs of drug packaging anddistribution.

9.2.1 Permuted Block DesignsThe permuted block schedule consists of a sequence of blocks that contain the treatmentassignments in desired ratios; the treatment assignments are randomly permuted withinthe blocks. Consider, for example, a study in which subjects are to be assigned to three

Page 228: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 215

treatments (Treatment 1, Treatment 2, and Treatment 3) using a 2 : 2 : 1 ratio. Thesmallest permuted block that provides the 2 : 2 : 1 treatment ratio is the block of size 5 andis a random permutation of

(1, 1, 2, 2, 3).

An example of a sequence of treatment assignments (with blocks bracketed to illustrate thetechnique) is

(2, 1, 1, 3, 2), (1, 1, 2, 3, 2), (3, 2, 1, 2, 1), . . . .

A larger block size (a multiple of 5) can also be used. If the block size of 10 is chosen, theblocks will each be a random permutation of

(1, 1, 1, 1, 2, 2, 2, 2, 3, 3).

Stratifying the randomization is easy with the permuted block schedule. Since any set ofblocks constitutes a permuted block schedule on its own, each stratum can be assigned aset of blocks from one common schedule. This property is useful in multi-center trialswhere, as recommended by the ICH Guidance entitled “Guidance on Statistical Principlesfor Clinical Trials” (ICH E-9), the allocation is typically stratified by center. Thestratification is implemented by assigning a set of blocks to each center.

The permuted block allocation provides a good balance in treatment assignments: whenmost of the blocks in the allocation schedule are filled, the treatment ratio of allocatedsubjects at the end of the study is close to the planned one. Unless there are many strataand thus, many unfilled blocks, the balance stays reasonably tight throughout theenrollment as well. This feature improves the efficiency of an interim analysis and helpsmitigate the accidental bias if a time trend in treatment effect is present.

These properties made the permuted block randomization an industry standard formasked clinical trials. For unmasked trials, where selection bias is an issue, otherapproaches such as a permuted block design with a random block size or maximalprocedure (Berger et al., 2003) might offer benefit.

9.2.2 Choice of Block Size in Permuted Block DesignsThe choice of block size for a masked permuted block schedule requires some consideration.It is preferred to have a block size big enough to include at least two subjects on eachtreatment to mitigate the impact of rare instances when a subject’s allocation may need tobe unblinded due to a serious adverse event. Having at least two subjects on eachtreatment within a permuted block will not narrow the list of possible assignments forother subjects from the block that the unblinded subject belongs to. This provision wouldlessen the potential for selection bias (if some of the subjects on the block are yet to beenrolled) and for observer bias.

This approach will allow using the minimal block size when the smallest block withrequired ratio already includes at least two subjects on each treatment (e.g., a 3 : 2 : 2allocation ratio). When the smallest block includes only one subject on any of thetreatments, e.g., a 2 : 2 : 1 ratio, the preferred block size will be at least twice as large asthe minimal block size (that is, at least 10).

The outlined block size selection strategy is intended to lessen the impact of unblindingand might not be advisable for a multi-center trial with small centers. For example, if theminimal block size is 5 and most of the centers are expected to enroll about six subjects,using a block size of 10 will lead to predominantly incomplete blocks and thus tosuboptimal balance in treatment assignments. A compromise can often be reached by usingone of the variations of permuted block designs (see Sections 9.3.2 and 9.3.4); however, if

Page 229: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

216 Pharmaceutical Statistics Using SAS: A Practical Guide

the possibility of unblinding is low, the most practical option might be to use the smallestblock size. In reality, designs with a block size of 2, in which an unblinding of one subjectleads to the unblinding of the other subject in the same block, are rarely used.

9.2.3 Generating Permuted Block SchedulesPermuted block schedules can be easily generated using PROC PLAN. PROC PLAN is apowerful procedure and often allows to generate the same randomization schedule usingseveral different sets of options. We will present one of the options in the example below.

EXAMPLE: Three-arm clinical trial with a 2 : 2 : 1 randomization ratioConsider a study in which 120 subjects are allocated to three treatment arms (Treatment1, Treatment 2, and Treatment 3) with a 2 : 2 : 1 ratio using the permuted blockrandomization with a block size 5. As is typically the case, it was decided to prepare anallocation schedule for more than 120 subjects by generating a total of 60 permuted blocks.

Program 9.1 uses PROC PLAN to generate a permuted block schedule for the three-armtrial. The first option (SEED=56789) in the PROC PLAN statement specifies a randomseed to make the schedule reproducible. If no seed is provided, the time of the day will beused as a default seed and each run of the program will result in a different schedule. Therandom seed used in the schedule should not be disclosed until the study is unblinded. Thenext statement

factors block=60 ordered treatment=5;

requests that the BLOCK variable be created in the output data set with values orderedfrom 1 to 60. For each value of BLOCK, five observations with five levels of the TREATvariable will be generated in a random order. Next,

output out=schedule treatment nvals=(1 1 2 2 3);

specifies the name of the output data set (SCHEDULE) as well as numeric values (1, 1, 2,2, and 3) that will be assigned to the five levels of the TREATMENT variable. PROCPLAN is followed by a DATA step that assigns consecutive allocation numbers (ANvariable) from 1 to 300 to all observations on the list. The treatment assignment is storedin the TREATMENT variable.

Program 9.1 Three-arm clinical trial with a 2 : 2 : 1 randomization ratio

ods listing close;proc plan seed=56789;

factors block=60 ordered treatment=5;output out=schedule treatment nvals=(1 1 2 2 3);

data schedule;set schedule;an=_n_;label treatment=’Treatment group’

block=’Block number’an=’Allocation number’;

proc print data=schedule noobs label;var an treatment block;ods listing;run;

Page 230: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 217

Partial output from Program 9.1

Allocation Treatment Blocknumber group number

1 3 12 2 13 2 14 1 15 1 16 1 27 1 28 2 29 3 210 2 2

Output 9.1 shows the first ten observations in the SCHEDULE data set. Theseobservations represent the allocation of the first ten subjects (two permuted blocks).

To generate 30 permuted blocks of size 10, the PROC PLAN options in Program 9.1need to be modified in the following way:

proc plan seed=56789;factors block=30 ordered treatment=10;output out=schedule treat nvals=(1 1 1 1 2 2 2 2 3 3);

The described approach to generating permuted block schedules relies on drawingpermuted blocks with replacement from a set of all existing permuted blocks with specifiedcontents. The resulting schedule does not necessarily use all existing permuted blocks of agiven structure and the same block can appear on the schedule multiple times. Thisapproach is simple to implement and, in most circumstances, adequately serves the needsof clinical researchers.

An alternative approach was described by Zelen (1974) for two-arm studies with a 1:1allocation that uses the permuted blocks of size 4. Zelen proposed to list all six existingpermuted blocks,

(1, 1, 2, 2), (1, 2, 1, 2), (1, 2, 2, 1), (2, 1, 1, 2), (2, 1, 2, 1), (2, 2, 1, 1),

and then randomly permute them to form a randomization sequence for the first 24subjects. The procedure can be repeated as many times as needed to produce therandomization schedule for any number of subjects. With this approach, the sets of allexisting permuted blocks are repeatedly used. This strategy can also be implemented withPROC PLAN. Section 9.3.4 describes the circumstances under which the Zelen approachcan be beneficial.

9.3 Variations of Permuted Block RandomizationAlthough permuted block randomization provides a satisfactory allocation for the majorityof clinical trials, some variations of it are also employed.

9.3.1 Permuted Block Design with a Variable Block SizeIn a study unmasked to the investigator, permuted block design with block of variable sizeis sometimes employed (ICH E-9), with the intention of making it harder for theinvestigator to guess the next treatment assignment and thus lessen the potential forselection bias. This strategy and its limitations are discussed in Rosenberger and Lachin

Page 231: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

218 Pharmaceutical Statistics Using SAS: A Practical Guide

(2002). They mention that, under a commonly used model for selection bias, variable blockallocation “yields a substantial potential for selection bias in an unmasked study that isapproximately equal to that associated with the average block size M”. On the other hand,variable block allocation reduces the expected number of assignments that may bepredicted with certainty compared to the sequence of permuted blocks of size M .

A permuted block randomization with variable block size can be generated in SAS usingrandom number generators.

EXAMPLE: A 1 : 1 randomization schedule with a variable block sizeConsider a clinical trial with 20 subjects who are to be allocated to two treatments in a 1:1ratio using permuted blocks of size 2, 4, or 6. The size of each permuted block will bechosen at random with probabilities 0.25, 0.5, and 0.25, respectively. To provide a schedulefor a minimum of 20 subjects, 10 permuted blocks need to be generated. The set of 10blocks can include up to 60 subjects; however, only the first 20 treatment assignments willbe used to allocate the study subjects.

Program 9.2 demonstrates how to generate a randomization schedule for the two-armtrial. First, the BLOCKS data set with 10 blocks of variable size is created. The size ofeach block is computed based on the HALF SIZE variable which is defined as follows

half_size=rantbl(12345,0.25,0.5,0.25);

This statement invokes the RANTBL function (with the random seed 12345) thatassigns values 1, 2, or 3 with probabilities 0.25, 0.5, and 0.25 to the HALF SIZE variable.After the block size is determined (it is equal to two times HALF SIZE since there are twotreatment groups), HALF SIZE observations are created in each treatment group withinthe block. After that, a random value uniformly distributed over (0,1) is added to the dataset (RANDOM variable) to randomly permute the treatment assignments within eachblock. The RANDOM variable is generated using the RANUNI function with the seed setto 678. The last DATA step assigns consecutive allocation numbers (AN variable) to allobservations on the list.

Program 9.2 A 1 : 1 randomization schedule with a variable block size

data blocks;do block=1 to 10;

half_size=rantbl(12345,0.25,0.5,0.25);do j=1 to half_size;

do treatment=1,2;random=ranuni(678);output;

end;end;

end;proc sort data=blocks out=schedule;

by block random;data schedule;

set schedule;an=_n_;label treatment=’Treatment group’

block=’Block number’an=’Allocation number’;

proc print data=schedule noobs label;var an treatment block;run;

Page 232: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 219

Output from Program 9.2

Allocation Treatment Blocknumber group number

1 2 12 1 13 1 14 2 15 1 26 1 27 2 28 2 29 2 310 1 311 1 312 2 313 1 414 2 415 2 416 1 417 1 418 2 419 2 520 1 521 2 522 1 523 2 624 1 625 1 726 2 727 2 728 1 729 2 830 1 831 1 932 2 933 2 934 1 935 1 1036 2 10

Output 9.2 displays the allocation schedule generated by Program 9.2. The scheduleuses the following sequence of block sizes

(4, 4, 4, 6, 4, 2, 4, 2, 4, 2).

It should be noted that, although in the example above, the first 20 subjects happenedto be split equally (10 and 10) between the two groups, an unbalanced allocation of 9 : 11might have also been an outcome. In general, when a sequence of permuted blocks with arandom block size is used, the maximum imbalance in treatment assignments between thetwo groups is determined by the largest block size allowed. In studies of small size, the needto achieve an exact balance in treatment assignments might be a consideration whenchoosing an allocation procedure.

Page 233: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

220 Pharmaceutical Statistics Using SAS: A Practical Guide

9.3.2 Constrained Block RandomizationClinical trials are often designed to have unequal allocation to different treatment arms.This might be based on efficiency considerations, ethical considerations (placing moresubjects on the treatment believed to be more efficient), the need to accumulate moreexperience with the experimental drug, or simply to meet regulatory exposurerequirements.

In multi-arm trials, the number of subjects enrolled in each arm may reflect the rolesplayed by the arms in the trial’s objective. For example, consider a three-arm study withan experimental drug arm, an active control arm and a placebo arm. The primaryobjective of the trial is to compare the experimental drug to the active control and theplacebo arm is included in the study to perform safety assessments. Power considerationsmay call for a size of 500 subjects in each of the two active treatment arms, and anadditional 200 subjects to be enrolled in the placebo group.

The allocation ratio of 5 : 5 : 2 will lead to a large block size (the smallest block size is12), which, in turn, can lead to logistical problems described below. However, changing thetreatment ratio to a similar, but more manageable ratio of 2 : 2 : 1, may not be acceptablesince it requires enrolling 50 additional placebo subjects.

When the allocation ratio calls for a large block size, permuted blocks can turn out tobe quite unbalanced with some treatment groups gathered at the beginning of the blockand other groups at the end. To illustrate, consider a three-arm clinical trial with a 5 : 5 : 2treatment ratio. With this ratio, the smallest block size is 12, and we can theoretically findthe following unbalanced block

(1, 1, 1, 1, 1, 2, 2, 2, 2, 2, 3, 3).

Highly unbalanced blocks of this kind are a concern in multi-center trials stratified bycenter. If some of the centers enroll less than a full block of subjects, these centers mightend up with an extreme imbalance in treatment assignments, or even without all treatmentarms represented. That may lead to problems at the analysis stage and, if there are manysmall centers, the described phenomenon will have a negative impact on the overall balanceof treatment assignments.

To avoid these problems when the block size is large, constrained randomizationmethods proposed by Youden (1964, 1972) can be used. Constrained block randomizationdoes not use every existing permuted block with a given treatment ratio but only thebetter balanced blocks specified in advance.

Consider again the trial with a 5 : 5 : 2 allocation to three treatments (Treatments 1, 2,and 3). Blocks of size 12 can be constructed in the following way: five permuted blocks(1, 2) or (2, 1) are lined up and then two Treatment 3 assignments are randomly inserted(one among the first six assignments and the other one among the last six assignments).An example of a block generated with the described algorithm is given below

(2, 1, 1, 2, 3, 1, 2, 2, 3, 1, 2, 1).

Another way to build a constrained randomization schedule with a block of size 12 is togenerate a random permutation of

(1, 1, 1, 2, 2, 3)

followed by a random permutation of

(1, 1, 2, 2, 2, 3),

or vice versa, at random. This set of assignments is less restrictive compared to theallocation algorithm described above.

Page 234: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 221

It is worth noting that other constrained block randomization algorithms have beenproposed in the literature. For example, Berger et al. (2003) introduced a randomizationprocedure (known as the maximal procedure) which uses any sequence in a block for whichthe maximum imbalance does not exceed a prespecified number.

With constrained randomization, blocks left incomplete at the end of enrollment willexhibit a better balance compared to a regular permuted blocks schedule, which might beespecially important if the trial uses interim analyses. Constrained randomization is alsoless susceptible to accidental bias caused by a time trend in a prognostic covariate.

Constrained randomization schedules can easily be generated using PROC PLAN byimposing a constraint on a full factorial design in which the number of factors is equal tothe block size (Song and Kuznetsova, 2003).

9.3.3 Allocation for Clinical Trials with Treatment Group SplittingA common example of constrained randomization—perhaps not recognized as such—ariseswhen treatment groups are split for a washout period or safety extension.

EXAMPLE: Dose-ranging study with a placebo washoutConsider a multi-center dose-ranging study with six treatment arms in which each arm isto be split in a 2 : 1 ratio at the end of the main study period for a placebo washout. Twothirds of each treatment arm will continue on the base study treatment, while theremaining third will be switched to placebo. This study design results in 12 differenttreatment regimens defined by a combination of a treatment arm (one of 6) and whether asubject will be switched to placebo or stay on the base study treatment during the placebowashout period.

At the base study start, the subjects are randomized to one of the 12 regimens (Arm 1without switch, Arm 1 with a switch, Arm 2 without switch, Arm 2 with a switch,. . ., Arm6 without switch, Arm 6 with a switch) in a

2 : 1 : 2 : 1 : 2 : 1 : 2 : 1 : 2 : 1 : 2 : 1

ratio. The minimal block size (18) can be too large for the study if most centers areexpected to enroll about 6 to 9 subjects. Therefore, a constrained randomization schedulewill be employed in which each block of 18 is built of three sub-blocks of six consecutiveallocations in the following way:• Each sub-block of six contains all six base study arms.• In addition, to evenly spread the allocations to placebo washout across the three

sub-blocks, two of the six base study arms within each sub-block are assigned to placebowashout and each sub-block has a different pair of the base study treatments assigned tothe placebo washout.

• The sub-blocks of six are distributed among the centers to achieve the balance acrossthe six base study arms within the centers and the balance in 12 treatment regimensacross the centers.

Program 9.3 implements the described constrained permuted block allocation algorithmand generates a schedule with ten blocks of size 18. First, PROC PLAN outputs ten blocks(BLOCK variable) of six arms (ARM variable assumes values 1 to 6 for each value ofBLOCK) into the WASHOUT data set. It assigns to each of the 6 arms a value of 1, 2, or 3(WASHOUT variable) by randomly permuting the set of values 1, 1, 2, 2, 3, and 3, amongthe six observations of each block:

Page 235: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

222 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 9.3 Dose-ranging trial with a placebo washout, Part 1

ods listing close;proc plan seed=56789;

factors block=10 ordered arm=6 ordered;treatments washout=6;output out=washout washout nvals=(1 1 2 2 3 3);

data washout;set washout;label block=’Block number’

arm=’Base study arm’washout=’Washout’;

proc print data=washout noobs label;ods listing;run;

Partial output from Program 9.3

BaseBlock studynumber arm Washout

1 1 31 2 21 3 31 4 11 5 21 6 12 1 12 2 22 3 32 4 22 5 32 6 13 1 23 2 13 3 23 4 33 5 13 6 34 1 24 2 14 3 24 4 34 5 34 6 1

Output 9.3 lists the first four blocks in the WASHOUT data set. Within each block, TheWASHOUT variable determines which of the six treatment arms will be switched toplacebo in the first, second or third sub-block of six, respectively. In the first block of 18(BLOCK=1), the first sub-block of six will have Arms 4 and 6 switched to placebo washout(WASHOUT=1), the second sub-block of six will have Arms 2 and 5 switched to placebowashout (WASHOUT=2) and the third sub-block of six will have Arms 1 and 3 switchedto placebo washout (WASHOUT=3).

The next step is to build the constrained blocks of 18 based on the WASHOUT data set.To do that, for each BLOCK=1 to 10, for each ARM=1 to 6 within the block, three

Page 236: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 223

observations (with the value of variable SUBBLOCK equal to 1, 2, and 3) are output intothe REGIMENS data set. The SWITCH variable is set to YES when theSUBBLOCK=WASHOUT (a sub-block where a respective arm switches to placebo) andNO otherwise. The treatment regimen is then defined as (2 ∗ ARM − 1) if the arm does notswitch to placebo and as 2 ∗ ARM if the arm switches to placebo. A random key(RANDOM) is added to each observation in order to randomly permute the treatmentregimens within each sub-block of 6. As in Program 9.2, this variable is uniformlydistributed over (0, 1).

Program 9.4 Dose-ranging trial with a placebo washout, Part 2

data regimens;set washout;length switch $3.;do subblock=1 to 3;

if washout=subblock then do;switch=’Yes’;treatment=arm*2;

end;else do;

switch=’No’;treatment=arm*2-1;

end;random=ranuni(6789);output;

end;label block=’Block number’

arm=’Base study arm’washout=’Washout’subblock=’Sub-block’switch=’Switch to placebo?’random=’Random key’treatment=’Treatment group’;

proc print data=regimens noobs label;var block arm washout subblock switch random treatment;run;

Partial output from Program 9.4

SwitchBlock Base study Sub- to Random Treatment

number arm Washout block placebo? key group

1 1 3 1 No 0.71089 11 1 3 2 No 0.93229 11 1 3 3 Yes 0.40721 21 2 2 1 No 0.05740 31 2 2 2 Yes 0.75990 41 2 2 3 No 0.42393 31 3 3 1 No 0.28561 51 3 3 2 No 0.65511 51 3 3 3 Yes 0.05164 61 4 1 1 Yes 0.92032 8

Output 9.4 lists the first ten observations in the REGIMENS data set.

Page 237: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

224 Pharmaceutical Statistics Using SAS: A Practical Guide

To complete the randomization schedule, the REGIMENS data set needs to be sorted byblock (BLOCK), subblock (SUBBLOCK) and randomly generated key (RANDOM) to havethe treatment regimens randomly permuted within each sub-block of 6 (see Program 9.5).

Program 9.5 Dose-ranging trial with a placebo washout, Part 3

proc sort data=regimens out=schedule;by block subblock random;

data schedule;set schedule;an=_n_;label an=’Allocation number’;

proc print data=schedule noobs label;var an arm switch block subblock treatment;run;

Partial output from Program 9.5

SwitchAllocation Base study to Block Sub- Treatmentnumber arm placebo? number block group

1 2 No 1 1 32 3 No 1 1 53 6 Yes 1 1 124 5 No 1 1 95 1 No 1 1 16 4 Yes 1 1 87 5 Yes 1 2 108 4 No 1 2 79 3 No 1 2 5

10 2 Yes 1 2 411 6 No 1 2 1112 1 No 1 2 113 3 Yes 1 3 614 5 No 1 3 915 1 Yes 1 3 216 2 No 1 3 317 4 No 1 3 718 6 No 1 3 1119 3 No 2 1 520 2 No 2 1 3

Output 9.5 lists the treatment assignments of the first 20 subjects in the dose-rangingtrial. The first column is the subject’s allocation number, the second column (Base studyarm) indicates the arm to which the subject will be assigned during the base study, and theswitch column shows whether or not the subject will be switched to placebo at the end ofthe base study. The column “Treatment group” contains the treatment regimen (one of 12)that the subject is allocated to at randomization. The other two columns help to explainthe allocation algorithm implemented in Programs 9.3, 9.4, and 9.5.

9.3.4 Balanced-Across-Centers AllocationA large number of incomplete blocks can seriously impair the balance of treatmentassignments in clinical trials with stratified randomization (Hallstrom and Davis, 1988). Asthe last block in each stratum is often left incomplete at the end of enrollment, stratified

Page 238: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 225

assignment can result in as many incomplete blocks as there are strata. If the number ofsubjects in each stratum is small the imbalance in treatment assignments might beconsiderable. Typically, this is of concern in a study with numerous small centers. Forexample, in a 30-center study stratified by center, as many as 30 blocks might beincomplete. Additional stratification by gender may result in as many as 2 × 30 = 60incomplete blocks.

To avoid having a large number of incomplete blocks, a central randomization algorithmcan be employed to allocate the subjects. With central randomization, a subject is assignedthe next available allocation number (and the respective treatment) in his or her stratum(for example, gender), regardless of what center this subject belongs to. This allocationscheme can be easily implemented with the Interactive Voice Response System (IVRS).However, if centers are small, the central randomization may result in some centers beingvery unbalanced in treatment assignments, up to a point of having only one treatmentassigned to all subjects at the center. That may lead to bias if a center happens to deviatefrom the other centers in efficacy or safety assessments due to a training issue. Also, it willlead to a suboptimal drug re-supply pattern, where the drug kits at the center with anunbalanced allocation will have to be replenished more frequently than would have beennecessary with a balanced allocation. Thus, even when an IVRS solution is available, thereare merits to having the allocation balanced within the centers.

If no IVRS or similar technology is available to support a central randomizationalgorithm in a study, the randomization is stratified by center because of drug distribution,if nothing else. In a study with a large number of small centers it may lead to a largenumber of incomplete blocks.

The imbalance caused by incomplete blocks can be lessened by having the blocksbalanced across the centers. Consider a study with 40 small centers and a 2 : 1 : 2 ratio ofallocation to Treatments 1, 2, and 3. The subject allocation is stratified by center tofacilitate drug distribution. Stratification by gender is desired but the small size of thecenters, which are expected to enroll six to nine subjects, makes it problematic. Almost allthe permuted blocks of five will likely be left incomplete.

To improve the balance in treatment assignments within each stratum, therandomization schedule for each stratum will be prepared following a modified version ofZelen’s (1974) approach described in Section 9.2.3. There are 30 different permutations of(1, 1, 2, 3, 3). These blocks can be arranged in six balanced sets of five blocks so that thefive permuted blocks within each set will have a 2 : 1 : 2 allocation ratio across the row ofsubjects allocated first, second, and so on. Thus, when the five blocks within the firstbalanced set are distributed among Centers 1 through 5 (see Table 9.1), a balance isestablished across these five centers among the subjects allocated first, second, and so on,at their respective center. If the same number of subjects are allocated to all five centers,there will be a perfect balance in treatment assignments across the centers even if all fiveblocks are incomplete. Of course, in real life the number of subjects enrolled will varyacross the centers but even in this case the described algorithm provides a better balance

Table 9.1 Permuted Blocks for the First Ten Centers in a Balanced-across-centers Allocation Scheme

CenterAllocation orderwithin each center 1 2 3 4 5 6 7 8 9 10

1st subject 2 1 3 1 3 2 3 1 1 32nd subject 3 2 1 3 1 3 1 3 1 23rd subject 1 3 3 1 2 1 1 2 3 34th subject 3 1 1 2 3 1 3 3 2 15th subject 1 3 2 3 1 3 2 1 3 1

Page 239: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

226 Pharmaceutical Statistics Using SAS: A Practical Guide

than an allocation schedule built from randomly selected permuted blocks. Further, thesecond balanced set of five blocks will be sent out to Centers 6 through 10; the procedurewill be repeated until all the blocks are generated for all 40 study centers.

The described randomization schedule can be generated in SAS (Song and Kuznetsova,2003). The algorithm is implemented in Program 9.6 using PROC PLAN. First, the sixbalanced sets of blocks are generated as six 5 × 5 Latin squares, where the value ofTREATMENT=1 is mapped to 2, and the vector of 4 remaining values (2, 3, 4, 5) ismapped to one of the six permutations of (1, 1, 3, 3). This is accomplished by calling PROCPLAN with numeric values for treatment specified as (21133), (21313), (21331), (23113),(23131) and (23311). After that, all six balanced sets of five blocks each are put together,the order of the sets is randomly permuted, and the order of the blocks within each set israndomly permuted.

Program 9.6 Balanced-across-centers allocation

proc plan seed=6767;factors row=5 col=5 ordered/noprint;treatments treatment=5 cyclic 4;output out=sq1 treatment nvals=(2 1 1 3 3);

proc plan seed=7878;factors row=5 col=5 ordered/noprint;treatments treatment=5 cyclic 4;output out=sq2 treatment nvals=(2 1 3 1 3);

proc plan seed=8989;factors row=5 col=5 ordered/noprint;treatments treatment=5 cyclic 4;output out=sq3 treatment nvals=(2 1 3 3 1);

proc plan seed=9797;factors row=5 col=5 ordered/noprint;treatments treatment=5 cyclic 4;output out=sq4 treatment nvals=(2 3 1 1 3);

proc plan seed=8791;factors row=5 col=5 ordered/noprint;treatments treatment=5 cyclic 4;output out=sq5 treatment nvals=(2 3 1 3 1);

proc plan seed=6917;factors row=5 col=5 ordered/noprint;treatments treatment=5 cyclic 4;output out=sq6 treatment nvals=(2 3 3 1 1);

data sixsq;set sq1(in=in1) sq2(in=in2) sq3(in=in3) sq4(in=in4) sq5(in=in5) sq6(in=in6);retain r1-r6;if _n_=1 then do;

r1=ranuni(4565);r2=ranuni(7271);r3=ranuni(8171);r4=ranuni(6171);r5=ranuni(7567);r6=ranuni(9231);

end;random=r1*in1+r2*in2+r3*in3+r4*in4+r5*in5+r6*in6;

proc sort data=sixsq out=schedule;by random row;

data schedule;set schedule;by random row;

Page 240: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 227

an=_n_;center=ceil(_n_/5);label an=’Allocation number’

center=’Center’treatment=’Treatment group’;

proc print data=schedule noobs label;var an center treatment;run;

Partial output from Program 9.6

Allocation Treatmentnumber Center group

1 1 22 1 33 1 14 1 35 1 16 2 17 2 28 2 39 2 110 2 311 3 312 3 113 3 314 3 115 3 216 4 117 4 318 4 119 4 220 4 3

Output 9.6 displays the treatment assignments of the first 20 subjects. It is easy to seethat the obtained assignments are identical to the assignments at Centers 1 through 4 inTable 9.1.

The benefits of balancing across the centers are most obvious when the block size islarge, e.g., in the case of a 5 : 5 : 2 allocation ratio. Balancing across centers can also beadvantageous when a stratification of the schedule by another factor, e.g., gender, iscontemplated in a study with small centers. Such a schedule will make the treatmentgroups better balanced in gender and will provide at least as good a balance in overalltreatment assignments as a randomization not stratified by gender.

In the extreme case of a very small stratum, e.g., when only two to three subjects percenter are expected to be enrolled in a study with a minimal block size of 8, the schedulebuilt of incomplete blocks balanced across the centers can be considered for the stratum.More examples can be found in Cho et al. (2004).

A balanced-across-centers allocation schedule is a special case (a single-factor case) ofthe factorial stratification proposed by Sedransk (1973). In the multi-factor case, thefactorial stratification provides a close balance across all first, second, third, and so onassignments in all strata. Factorial stratification is most beneficial if subjects are evenlydistributed across the strata, which is often not the case.

Page 241: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

228 Pharmaceutical Statistics Using SAS: A Practical Guide

9.4 Allocations Balanced on Baseline CovariatesWhen certain subject characteristics (baseline covariates) are known to affect the response,it is often important to have the treatment groups balanced with respect to thesecovariates:

• A balanced allocation helps minimize bias due to covariate imbalances unaccounted forin the analysis model.

• A balance with respect to important covariates improves the efficiency of the analysis(Senn, 1997). Note, however, that in large trials the gain in power resulting from the useof a balanced allocation is typically small compared to post-stratified analysis withoutbalanced allocation (McEntegart, 2003). At the same time, as pointed out byMcEntegart, even a large trial can suffer from a substantial loss of power due to anunbalanced allocation in the presence of many small strata. Also, an allocation balancedby baseline predictors can considerably improve the efficiency of interim and subgroupanalyses.

• The results of a trial with treatment groups balanced with respect to major covariatesare more convincing for the medical community. Serious imbalance in importantcovariates might raise concerns even if it is adjusted for in the analysis.

It is debatable to what extent the balance in prognostic factors should be pursued, whenstatistical analysis can account for any imbalances in covariates; see, for example, a heateddiscussion following Atkinson (1999). The decision to balance or not to balance in aparticular study can be guided by an assessment of the probability to conclude with animbalance that is either perceived as dangerously high in the clinical community or exceedsthe boundaries within which the model assumptions can be trusted. More insights can begained from McEntegart (2003).

It is recommended that the baseline predictors balanced upon be included in theanalysis model (International Conference on Harmonization, 1998; Gail, 1988; Simon, 1979;Kalish and Begg, 1987; Senn, 1997; Scott et al., 2002). A failure to do so might result in anincorrect Type I error rate. When the randomization-based analysis is performed, it shouldfollow the randomization procedure (Rosenberger and Lachin, 2002).

9.4.1 Stratified Permuted Block RandomizationThe most common way to achieve balance in a given factor is stratified randomizationwhich relies on creating a separate randomization schedule for each level of the factor. Ifthere are several baseline covariates to balance upon, a separate restricted randomizationschedule (most commonly, a permuted block schedule) is prepared for each stratificationcell defined by a combination of factor levels. For example, in a study balanced by gender(male or female) and smoking status (smoker, ex-smoker or never-smoker), a separateschedule is generated for each of the six strata formed by a combination of the factor levels.

The stratified randomization approach has its limitations: only a small number offactors can be balanced upon. If the number of strata is large, some strata will have few orno subjects resulting in an inadequate balance across factor levels (Therneau, 1993;Rosenberger and Lachin, 2002; Hallstrom and Davis, 1988).

9.4.2 Covariate-Adaptive Allocation ProceduresWhen there are too many important prognostic factors for stratification to handle, one ofthe covariate-adaptive allocation procedures can be used to provide a balance in selectedcovariates. Such procedures are dynamic in nature—the treatment assignment of a subjectdepends on the subject’s vector of covariates and thus is determined only when the subject

Page 242: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 229

arrives. It is conceptually different from the regular stratified randomization method thatrelies on a fixed allocation schedule prepared for each stratum prior to the study start.

Minimization, pioneered by Taves (1974) and expanded by Pocock and Simon (1975), isthe most commonly used covariate-adaptive allocation procedure. Minimization produces amarginal balance in each individual factor but not in individual factor interaction cells. Forexample, when balancing on gender and smoking status, the balance in treatmentassignments is achieved across factor levels (across males, females, smokers, ex-smokers,and never-smokers) but not within each cell, e.g., male smokers, as is the case with thestratified randomization. Because of that, minimization can simultaneously be balanced ona large number of factors even in a moderate size trial.

Minimization achieves the balance in treatment assignments across factor levels bychoosing the allocation for the new subjects that would result in the least possibleimbalance (in some sense) across the set of his or her baseline characteristics.

In what follows we will describe the minimization algorithm proposed by Taves with avariance imbalance function popularized by Freedman and White (1976). Due to itssimplicity, this algorithm is frequently used in clinical trials (McEntegart, 2003) and isoften described in the literature (Senn, 1997; Scott et al., 2002).

9.4.3 Taves Minimization AlgorithmConsider a parallel study in which subjects are to be allocated equally to two treatmentgroups (Treatment A and Treatment B) and the allocation needs to be balanced in gender(male or female) and smoking status (smoker, ex-smoker, or never-smoker).

When the minimization algorithm is described, it is convenient to assign scores of 1 and−1 to the treatment groups A and B, respectively. Suppose a male ex-smoker arrives forallocation when there are 20 allocated subjects in the trial. To select the treatmentassignment for this subject, we count the number of males and the number of ex-smokers ineach of the treatment arms. Assume that there are five males in the Treatment A groupand seven males in the Treatment B group. The imbalance across males, defined as thedifference in number of males allocated to A versus B, is 5 − 7 = −2. Also, there are threeex-smokers in the Treatment A group and two ex-smokers in the Treatment B group. Theimbalance across ex-smokers is 3 − 2 = 1. The total imbalance, defined as the sum ofimbalances across males and across ex-smokers, is (−2) + 1 = −1. The negative totalimbalance indicates that, overall, Treatment A is underrepresented among males andamong ex-smokers combined. Thus, to improve the balance, the male ex-smoker isallocated to Treatment A. When the group totals are equal, the subject is allocated to oneof the treatment arms at random with equal probability.

Given the covariates and treatment allocations of the subjects already in the study, thetreatment assignment of a new subject is fully determined by his or her set of covariates,except when a tie in group totals is encountered. This is why Scott et al. (2002) referred tothe Taves minimization algorithm as “largely nonrandom”—that is, deterministicassignments occur more often than random ones. Nevertheless, simulations show thatassignments at random occur often enough to provide a reasonably rich set of possibleallocation sequences for a given sequence of covariates (Kuznetsova and Troxell, 2004).Thus, in a masked trial, a largely deterministic nature of the Taves minimization algorithmdoes not lead to selection bias. However, in a single-center unmasked trial, a large share ofthe treatment assignments will be predictable, providing a considerable opportunity forselection bias.

9.4.4 Pocock-Simon Minimization AlgorithmPocock and Simon (1975) extended the Taves minimization algorithm to make treatmentassignments less predictable. This is achieved by using an additional random element ateach treatment assignment. A subject is allocated to the treatment that results in the least

Page 243: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

230 Pharmaceutical Statistics Using SAS: A Practical Guide

imbalance with probability p < 1 rather than p = 1. If p is close to 1 (p = 0.9 or p = 0.95are often used) the Pocock-Simon procedure still has good balancing properties andsomewhat less potential for selection bias in an unmasked trial. The ICH E-9 guidancerecommends using a random element at each allocation.

To define the Pocock-Simon minimization algorithm, consider a clinical study withsubjects allocated in a 1 : 1 ratio to Treatments A and B. The allocation scheme in thisstudy needs to be balanced by gender (male or female) and the smoking status (smoker,ex-smoker, or never-smoker). As was explained above, the Pocock-Simon algorithm extendsthe Taves algorithm by introducing a random element to each allocation step. In this trial,a subject will be assigned to the treatment that results in a smaller imbalance withprobability p = 0.9 and to the opposite treatment with probability p = 0.1. When the tie intotal imbalances is encountered, the treatment will be assigned by a toss of a fair coin.

The Pocock-Simon procedure, which can be expanded to more than two treatmentgroups, allows the use of different measures of imbalance across a factor level. If some ofthe factors are considered more important than others, they can be included in combinedimbalance with higher weight. If an interaction between the two factors is known to affectthe response, the interaction should be included as a factor in the minimization algorithm(Pocock and Simon, 1975).

The minimization approach has been shown to provide a good marginal balance in alarge number of factors simultaneously (Taves, 1974; Pocock and Simon, 1975; Therneau,1993; Begg and Iglewicz, 1980; Birkett, 1985; Zielhuis et al., 1990; Weir and Lees, 2003).By McEntegart’s (2003) estimate, minimization has been used in more than 1,000 trials,including several prestigious mega-trials.

9.4.5 Implementation of Minimization Algorithms in SASBelow we describe the %ASSIGN macro that must be invoked each time a new subject isavailable for allocation. The set of the subject’s covariates is specified through the macroparameters. The macro assigns a treatment to the new subject and also updates theALL ANS data set that stores the allocation numbers, covariates, and treatmentassignments of all allocated study subjects. This data set will have one observation for eachstudy subject and will include the following variables:

• The study subjects will be identified by their allocation numbers (AN variable),assigned to them in the order they were allocated.

• The treatment assignments of the study subjects will be stored in the TREATMENTvariable. Treatments A and B will be coded by 1 and −1, respectively.

• The set of covariates of each subject will be described by five 0/1 variables, C1 to C5.The first three variables, C1, C2, and C3, are 0/1 indicators of the subject’s level of thesmoking status (smoker, ex-smoker, or never-smoker, respectively), while C4 and C5 arethe indicators of the subject’s gender (male or female, respectively). For example, a maleex-smoker will have the following set of variables: C1=0, C2=1, C3=0, C4=1, C5=0.

• Lastly, there will be five variables, M1 to M5, to store the marginal imbalances, that is,the differences in the number of subjects allocated to A versus B across smokers (M1),ex-smokers (M2), never-smoker (M3), males (M4), and females (M5), respectively, thatresult after the subject is allocated.

Before the first subject is allocated, the ALL ANS data set must be initialized bysetting all of the variables to 0 in the following step as shown in Program 9.7.

Page 244: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 231

Program 9.7 Initialize the parameters in the ALL ANS data set

data all_ans;label an=’Allocation Number’

c1=’Smoker’c2=’Ex-smoker’c3=’Never-smoker’c4=’Male’c5=’Female’m1=’Imbalance across Smokers’m2=’Imbalance across Ex-smokers’m3=’Imbalance across Never-smokers’m4=’Imbalance across Males’m5=’Imbalance across Females’treatment=’Treatment’;

input an c1-c5 m1-m5 treatment;datalines;0 0 0 0 0 0 0 0 0 0 0 0 0;run;

Each time a new subject arrives for allocation, the %ASSIGN macro is called to determinethe treatment assignment of the new subject and update the ALL ANS data set. Themacro works in the following way:

• The macro reads the current marginal imbalances into a one-observation data set(ASSIGN data set). It creates indicator variables C1 to C5 that describe the covariatesof the new subject and assigns them values of the macro parameters &C1 to &C5. Thelast macro parameter, &P, is the probability of assigning a new subject to Treatment B.

• The treatment assignment for the new subject is determined by the scalar product ofthe vectors (C1, C2, C3, C4, C5) and (M1, M2, M3, M4, M5), the so-called totalimbalance (TOTIMB variable). If TOTIMB=0, the TREATMENT variable is set to 1or −1 with probability 0.5. If TOTIMB is positive, the TREATMENT variable is set to−1 (Treatment B) with probability &P and 1 (Treatment A) with probability 1-&P. IfTOTIMB is negative, TREATMENT=1 (Treatment A) with probability &P and valueTREATMENT=−1 (Treatment B) with probability 1 to &P.

• After the new subject has been assigned to a treatment group, the marginal imbalancesM1 to M5 are updated in the ASSIGN data set. This data set, which contains theallocation number, covariate indicators C1 to C5, treatment assignment for the newsubject, and updated marginal imbalances, is appended to the ALL ANS data set.

We need to go through these steps every time a subject arrives for allocation. The%ASSIGN macro is defined in Program 9.8.

Program 9.8 The %ASSIGN macro

%macro assign(c1,c2,c3,c4,c5,p);data assign;

set all_ans(keep=m1-m5 an) end=lastobs;if lastobs;c1=&c1; c2=&c2; c3=&c3; c4=&c4; c5=&c5;totimb=c1*m1+c2*m2+c3*m3+c4*m4+c5*m5;* Assign -1 or 1 with probability 0.5 if totimb=0;if totimb=0 then treatment=2*rantbl(0,0.5,0.5)-3;

Page 245: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

232 Pharmaceutical Statistics Using SAS: A Practical Guide

* Assign 1 with probability &p and -1 with probability 1-&p if totimb>0,otherwise assign -1 with probability &p and 1 with probability 1-&p;

else treatment=-sign(totimb)*(2*rantbl(0,1-&p,&p)-3);an=an+1;m1=m1+c1*treatment;m2=m2+c2*treatment;m3=m3+c3*treatment;m4=m4+c4*treatment;m5=m5+c5*treatment;keep an c1-c5 m1-m5 treatment;

data all_ans;set all_ans assign;if an>0;run;

%mend assign;

Program 9.9 invokes the %ASSIGN macro to allocate the first three subjects in a studythat involves a never-smoking male, never-smoking female, and a smoker male in the orderof arrival. These subjects will be assigned to the treatment that produces better balancewith probability 0.9 and thus &P=0.9.

Program 9.9 Pocock-Simon minimization algorithm with p = 0.9

* 1st subject: never-smoking male;%assign(c1=0,c2=0,c3=1,c4=1,c5=0,p=0.9);* 2nd subject: never-smoking female;%assign(c1=0,c2=0,c3=1,c4=0,c5=1,p=0.9);* 3rd subject: smoker male;%assign(c1=1,c2=0,c3=0,c4=1,c5=0,p=0.9);proc print data=all_ans noobs;

var an treatment c1-c5 m1-m5;run;

Output from Program 9.9

an treatment c1 c2 c3 c4 c5 m1 m2 m3 m4 m5

1 -1 0 0 1 1 0 0 0 -1 -1 02 1 0 0 1 0 1 0 0 0 -1 13 1 1 0 0 1 0 1 0 0 0 1

Output 9.9 lists the allocation numbers, treatment assignments, and values of the C1 toC5 and M1 to M5 variables. The three subjects were assigned to Treatments B, A, and A,respectively.

To change the probability of assigning subjects to the treatment that produces betterbalance, we must change the &P macro parameter. For example, increasing the value of&P to 0.95 will result in a tighter balance with respect to baseline covariates.

To implement the Taves minimization algorithm, the &P macro parameter is set to 1.There are other approaches to balancing an allocation on baseline covariates. Atkinson

(1982) proposed an approach based on optimal design considerations that focuses onminimizing the variance of treatment contrasts in the presence of covariates rather than onbalancing over the covariates to minimize the bias. A different approach (Miettinen, 1976)is based on stratifying by a single risk score that accounts for the effect of all knowncovariates.

Page 246: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 233

The CPMP Points to Consider document on adjustment for baseline covariates (EAEMPCPMP, 2003) discourages the use of covariate-adaptive allocation procedures. The issuesinvolved are discussed by Roes (2004). He shows that some of the arguments againstdynamic allocation procedures (e.g., predictability of the assignments) apply to stratifiedrandomization as well, and might be even more pronounced with stratified randomization.The utility of dynamic allocation procedures is well described by McEntergart (2003).

9.5 SummaryIn this chapter we describe randomization procedures used by the pharmaceutical industry.We devote most attention to the popular permuted block design and its variations broughtin by variable block size, constrained randomization, and randomization, balanced acrossthe centers. For each of the described approaches, we provide SAS code to create arandomization schedule.

We also discuss covariate-adaptive allocation procedures. When there is a need tobalance the treatment groups with respect to baseline covariates, the stratified permutedblock randomization is typically used. However, if the number of baseline covariates tobalance upon is large, stratified randomization might not be feasible. In this case, one ofthe dynamic covariate-adaptive allocation procedures can be used to achieve the requiredbalance. We describe the popular minimization procedure—a largely deterministic Taves(1974) version and a version by Pocock and Simon (1975) where a random element is addedat each allocation step. Both algorithms are implemented in a SAS macro. An examplethat illustrates the use of the macro to randomize the patients dynamically is provided.

The randomization designs not covered in this chapter include biased coin designs(Efron, 1971), the maximal procedure (Berger et al., 2003), and designs based on urnmodels (see the review in Wei and Lachin, 1988). The maximal procedure (Berger et al.,2003) was proposed as an alternative to a sequence of permuted blocks of small sizes. Themaximal procedure maintains an imbalance no larger than a prespecified positive integernumber b throughout the enrollment and provides less potential for selection bias than theset of randomized blocks of size 2b. Although originally developed for trials with twotreatments and a 1 : 1 treatment ratio, the maximal procedure can be easily expanded formore than two treatments. Each of these designs achieves a certain trade-off between thetreatment balance and the amount of randomization the design provides.

Randomization designs can be viewed as adaptive allocations that use previoustreatment assignments to modify the probability of the next assignment to ensure goodbalance in treatment assignments and to provide some amount of randomization. Acompletely different class of randomization designs are response-adaptive allocationprocedures. These designs use responses observed so far in the trial to change allocationaway from perfectly balanced allocation based on a certain objective. We refer the readerto Hu and Ivanova (2004) for the most recent review of response adaptive designs.

AcknowledgmentsWe gratefully acknowledge advice and helpful interactions with Deborah Shapiro, LarryGould, Meehyung Cho, Inna Perevozskaya, Chau Thach, Cindy Song, and John Troxell ofMerck and Co., Inc., with whom the first author has collaborated on randomization issuesover the years.

ReferencesAtkinson, A.C. (1982). “Optimum biased coin designs for sequential clinical trials with prognostic

factors.” Biometrics. 69, 61–67.

Page 247: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

234 Pharmaceutical Statistics Using SAS: A Practical Guide

Atkinson, A.C. (1999). “Optimum biased-coin designs for sequential treatment allocation withcovariate information with Discussion (1999).” Statistics in Medicine. 18, 1741–1755.

Begg, C. B., Iglewicz, B. (1980). “A treatment allocation procedure for sequential clinical trials.”Biometrics. 36, 81–90.

Berger, V.W., Ivanova, A., Knoll, M. (2003). “Minimizing Predictability while Retaining Balancethrough the Use of Less Restrictive Randomization Procedures.” Statistics in Medicine. 22,3017–3028.

Birkett, N. J. (1985). “Adaptive allocation in randomized controlled trials.” Controlled ClinicalTrials. 6, 146–155.

Blackwell, D., Hodges, J. Jr. (1957). “Design for the control of selection bias.” Annals ofMathematical Statistics. 28, 449–460.

Cho, M., Kuznetsova, O., Perevozskaya, I., Thach, C. (2004). Allocation Procedures in ClinicalTrials Globally and Locally. Merck Technical Report, 101.

Committee for Proprietary Medicinal Products. (2003). “Points to consider on adjustment forbaseline covariates.” The European Agency for Evaluation of Medicinal Products.CPMP/EWP/2863/99.

Efron, B. (1971). “Forcing a sequential experiment to be balanced.” Biometrika. 58, 403–417.Follmann, D., Proschan, M. (1994). “The effect of estimation and biasing strategies on selection bias

in clinical trials with permuted blocks.” Journal of Statistical Planning and Inference. 39, 1–17.Freedman, L. S., White, S. J. (1976). “On the use of Pocock and Simon’s method for balancing

treatment numbers over prognostic factors in the controlled clinical trial.” Biometrics. 32,691–694.

Gail, M. N. (1988). “The effect of pooling across strata in perfectly balanced studies.” Biometrics.44, 151–163.

Hallstrom, A., Davis, K. (1988). “Imbalance in treatment assignments in stratified blockedrandomization.” Controlled Clinical Trials. 9, 375–382.

Hu, F., Ivanova, A. (2004). “Adaptive design.” Encyclopedia of Biopharmaceutical Statistics. 2d ed.New York: Marcel Dekker.

International Conference on Harmonization (ICH) (1998). “Guidance on Statistical Principles forClinical Trials.” Federal Register. International Conference on Harmonization, E-9 Document.63, 179, 49583–49598.

Kalish, L. A., Begg, C. B. (1987). “The impact of treatment allocation procedures on nominalsignificance levels and bias.” Controlled Clinical Trials. 8, 121–135.

Kuznetsova, O.M., Troxell, J. K. (2004). “Maximum imbalance in treatment assignments in aclinical trial that uses the minimization allocation procedure.” 2004 Proceedings of the AmericanStatistical Association. Biopharmaceutical Section.

McEntegart, D. J. (2003). “The pursuit of balance using stratified and dynamic randomizationtechniques: An overview.” Drug Information Journal. 37, 293–308.

Miettinen, O. S. (1976). “Stratification by a multivariate confounder score.” American Journal ofEpidemiology. 104, 609–620.

Pocock, S. J., Simon, R. (1975). “Sequential treatment assignment with balancing for prognosticfactors in the controlled clinical trials.” Biometrics. 31, 103–115.

Roes, Kit C.B. (2004). “Regulatory Perspectives: Dynamic allocation as a balancing act.”Pharmaceutical Statistics. 3, 187–191.

Rosenberger, W.F., Lachin, J.M. (2002). Randomization in Clinical Trials: Theory and Practice.New York: Wiley.

Scott, N. W., McPherson G.C., Ramsay, C.R., Campbell. M.K. (2002). “The method ofminimization for allocation to clinical trials: a review.” Controlled Clinical Trials. 23, 662–674.

Sedransk, N. (1973). “Allocation of sequentially available units to treatment groups.” Proceedings ofthe 39th International Statistical Institute. Book 2, 393–400.

Senn, S. (1997). Statistical Issues in Drug Development. Chichester: Wiley.

Page 248: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 9 Allocation in Randomized Clinical Trials 235

Simon, R. (1979). “Restricted randomization designs in clinical trials.” Biometrics. 35, 503–512.Song, C., Kuznetsova, O.M. (2003). “Implementing Constrained or Balanced-Across-the-Centers

Randomization with SAS v8 Procedure PLAN.” Proceedings of the PharmaSUG AnnualMeeting, 473–479.

Taves, D. R. (1974). “Minimization: A new method of assigning patients to treatment and controlgroups.” Clinical Pharmacology Therapeutics. 15, 443–453.

Therneau, T. M. (1993). “How many stratification factors are ‘too many’ to use in a randomizationplan?” Controlled Clinical Trials. 14, 98–108.

Wei, L.J. (1978). “An application of an urn model to the design of sequential controlled clinicaltrials.” Journal of the American Statistical Association. 73, 559–563.

Wei, L.J., Lachin, J.M. (1988). “Properties of the urn randomization in clinical trials.” ControlledClinical Trials. 9, 345–364.

Weir, C. J., Lees, K. R. (2003). “Comparison of stratification and adaptive methods for treatmentallocation in an acute stroke clinical trial.” Statistics in Medicine. 22, 705–726.

Youden, W. J. (1964). “Inadmissible random assignments.” Technometrics. 6, 103–104.Youden, W. J. (1972). “Randomization and experimentation.” Technometrics. 14, 13–22.Zelen, M. (1974). “The randomization and stratification of patients to clinical trials.” Journal of

Chronic Disease. 27, 365–375.Zielhuis, G. A. , Straatman, H., Hof-Grootenboer, A. E. van ’T, van Lier, H. J. J., Rach, G. H., van

den Broek, P. (1990). “The choice of a balanced allocation method for a clinical trial in otitismedia with effusion.” Statistics in Medicine. 9, 237–246.

Page 249: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 250: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10

Sample-Size Analysis forTraditional HypothesisTesting: Concepts and Issues

Ralph G. O’BrienJohn Castelloe

10.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238

10.2 Research Question 1: Does “QCA” Decrease Mortality in Children with Severe

Malaria? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 240

10.3 p-Values, α, β and Power . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241

10.4 A Classical Power Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 243

10.5 Beyond α and β: Crucial Type I and Type II Error Rates . . . . . . . . . . . . . . . . . . . . . . . . . 249

10.6 Research Question 1, Continued: Crucial Error Rates for Mortality Analysis . . . . . . . . 251

10.7 Research Question 2: Does “QCA” Affect the “Elysemine : Elysemate”

Ratios (EER)?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .253

10.8 Crucial Error Rates When the Null Hypothesis Is Likely to Be True . . . . . . . . . . . . . . . 262

10.9 Table of Crucial Error Rates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263

10.10 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264

Appendix A Guidelines for “Statistical Considerations” Sections . . . . . . . . . . . . . . . . . . . . . . . 264

Appendix B SAS Macro Code to Automate the Programming . . . . . . . . . . . . . . . . . . . . . . . . . 265

Sample-size analysis continues to be transformed by ever-improving strategies, methods,and software. Using these tools intelligently depends on what the investigators understandabout statistical science and what they know and conjecture about the particular researchquestions driving the study planning. This chapter covers only the most common type ofsample-size analysis—power analysis, i.e., studying the chance that a given hypothesis testwill be “statistically significant,” p ≤ α. We focus on the core concepts and issues that thecollaborating statistician must master and that key investigators must understand.

Ralph O’Brien is Professor, Quantitative Health Sciences, Cleveland Clinic, USA ([email protected]). John Castelloeis Senior Research Scientist, SAS Institute, USA ([email protected]).

Page 251: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

238 Pharmaceutical Statistics Using SAS: A Practical Guide

We begin by reviewing p values and discuss how to conduct sample-size analyses thatfocus on the classical Type I and Type II error rates, α and β. Then we go further toconsider two other error rates, the crucial Type I error rate, α∗, which is the chance thatthe null hypothesis is true even though p ≤ α, and the crucial Type II error rate, β∗, definedas the chance that the null hypothesis is false in some particular way even though p > α.We argue that α∗ and β∗ are just as relevant (if not more so) than α and β. These issuesare explored in depth through two examples stemming from a straightforward clinical trial.

10.1 IntroductionIn their “Perspectives on Large-Scale Cardiovascular Clinical Trials for the NewMillennium,” Dr. Eric Topol and colleagues (1997) provide a fine preamble to ourdiscussions:

The calculation and justification of sample size is at the crux of the design of a trial. Ideally,clinical trials should have adequate power, ≈ 90%, to detect a clinically relevant differencebetween the experimental and control therapies. Unfortunately, the power of clinical trials isfrequently influenced by budgetary concerns as well as pure biostatistical principles. Yet anunderpowered trial is, by definition, unlikely to demonstrate a difference between theinterventions assessed and may ultimately be considered of little or no clinical value. From anethical standpoint, an underpowered trial may put patients needlessly at risk of a new therapywithout being able to come to a clear conclusion.

In addition, it must be stressed that investigators do not plan studies in a vacuum. Theydesign them based on their knowledge and thoughtful conjectures about the subjectmatter, on results from previous studies, and on sheer speculation. They may already befar along in answering a research question, or they may be only beginning. RichardFeynman, the 1965 Nobel Laureate in Physics and self-described “curious character,”stated this somewhat poetically (1999, P. 146):

Scientific knowledge is a body of statements of varying degrees of uncertainty,some mostly unsure,

some nearly sure,none absolutely certain.

This reflects what we will call The March of Science, which for clinical research issketched in Figure 10.1.

As we step forward, our sample-size considerations need to reflect what we know. At anypoint, but especially at the beginning, the curious character inside us should be free toconduct observational, exploratory, or pilot studies because, as Feynman said, “somethingwonderful can come from them.” Such studies are still “scientific” but they are forgenerating new and more specific hypotheses, not for testing them. Accordingly, little or noformal sample-size analyses may be called for. But to become “nearly sure” about ouranswers, we typically conduct convincing confirmatory studies under specific protocols.This often requires innovative and sophisticated statistical planning, which is usuallyheavily scrutinized by all concerned, especially by the reviewers. No protocol is everperfect, but paraphrasing the New York Yankee catcher and populist sage, Yogi Berra,Don’t make the wrong mistake.

Medical research is still dominated by traditional (frequentist) hypothesis testing andclassical power analysis. Here, investigators and reviewers typically ask, “What is thechance (inferential power) that some given key p-value will be significant, i.e. less thansome specified Type I error rate, α?” Thus, we cannot understand inferential powerwithout knowing what p-values are and what they are not. Researchers rely on them to

Page 252: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 239

Figure 10.1 March of science in clinical research

�������

��

�����

���

����

����

���

����

���

���

����

���

��� ��

���

���

�����

���

����

������

���

���

���������� ������

�������� ��� ��� ����

��� ���������� ����

��� ���� ������� ����

���������������� ��������� ���� ��� ����������� ����� ��

�������������� ����� �� ��� ��������������������������� ���������� ������� ���

����� �� �������������� ������ ���������� ������� ���

������������ �

assess whether a given null hypothesis is true, but p-values are random variables, so theycan mislead us into making Type I and II errors. The respective classical error rates arecalled α and β = 1 − power. All of this is reviewed in detail.

This chapter also considers other error rates that relate directly to two crucial questionsthat researchers should address. First, if a test turns out to be significant, what is thechance that its null hypothesis is actually true (Type I error)? A great many researchersthink that this chance is at most α. They might say something like, “We will use α = 0.05as our level for statistical significance, so if we get a significant result, we will be more than95% confident that the treatments are different with respect to this outcome.” Researcherswant to be able to make statements like this, but this particular logic is wrong. Likewise, if atest turns out to be non-significant, they might ask, “What is the chance that its nullhypothesis is actually false (Type II error) to some particular degree?” Many researchersthink this is the usual Type II error rate, β. It is not.

So, what is an appropriate way to do this? We describe something we call the crucialType I error rate (here, α∗), which is the chance that the null hypothesis is true even afterobtaining significance, p ≤ α. Similarly, the crucial Type II error rate (β∗) is the chancethat the null hypothesis is false in some particular way even though a p > α result hasoccurred. We argue that α∗ and β∗ are just as relevant (if not more so) than α and β. Wedemonstrate how crucial error rates can be guesstimated if investigators are willing to stateand justify their current belief about the chance that the null hypothesis is indeed false.Importantly, for a given α level, greater inferential power reduces both crucial error rates.

All these concepts will be developed and illustrated by carrying out a sample-sizeanalysis for a basic two-group trial to compare two treatments for children with severemalaria: usual care only versus giving an adjuvant drug known to reduce high levels oflactic acid. Two planned analyses will be covered. The first compares the groups withrespect to a binary outcome, death within the first ten days. The second compares them ona continuous outcome, the ratio of two amino acids measured in plasma, using baselinevalues as covariates. The principles covered apply to any traditional statistical test beingused to try to reject a null hypothesis, including analyses far more complex than thosediscussed here.

Page 253: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

240 Pharmaceutical Statistics Using SAS: A Practical Guide

While obtaining an appropriate and justifiable sample size is important, going throughthe analytical process itself may be just as vital in that it forces the research team to workcollaboratively with the statistician to delineate and critique the rationale undergirding thestudy and all the components of the research protocol. The investigators must specify tightresearch questions, the specific research design, the various measures, and an analysis plan.They must come to agree on and justify reasonable conjectures for what the “infinitedataset” may be for their study. In essence, they must imagine how the entire study willproceed before the first subject is recruited. The “group think” on this can be invaluable.

Our reader audience includes both collaborating statisticians and content investigators.While the examples given here involve clinical trials, the principles apply broadly across allof science. Therefore, we present almost no mathematical details.

The SAS procedures POWER and GLMPOWER are the primary computationalengines, but we use only a small portion of their capabilities. Far more information can befound in the current SAS/STAT User’s Guide.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

10.2 Research Question 1: Does “QCA” Decrease Mortality in Childrenwith Severe Malaria?According to a report released in 2003 by the World Health Organization, malaria remainsone of the world’s foremost health problems, killing at least one million people annually,mostly children under five years old in sub-Saharan Africa. Lactic acidosis (toxic levels oflactic acid in the blood) is a frequent complication in severe malaria and is an incrementalstatistical predictor (“independent risk factor”) of death. Moreover, a plausible biologicalrationale supports the hypothesis that lactic acidosis is a contributing cause of death.

Dr. Peter Stacpoole of the University of Florida has spent decades investigating thesafety and efficacy of dichloroacetate (DCA) for treating lactic acidosis in genetic andacquired diseases. In 1997–99, he collaborated with Dr. Sanjeev Krishna of the Universityof London to lead a team that conducted a small, randomized, double-blind, controlled trialof quinine-only versus quinine+DCA in treating lactic acidosis in Ghanaian children withsevere malaria (Agbenyega et al., 2003). They concluded that a single infusion of DCA waswell-tolerated, did not appear to interfere with quinine and, as hypothesized, reduced bloodlactate levels. The sample size of N = 62 + 62 was much too small to support comparingmortality rates. The authors concluded that a large prospective study was warranted.

From now on the story is fictionalized. Suppose “quadchloroacetate” (QCA) has thesame molecular structure as DCA at the active biological site, and has now been shown inlarge animal and human studies to be clinically equivalent to DCA in quickly reducingabnormally high blood lactate levels. However, QCA is less expensive to produce (aboutUS$1/dose) and has a longer shelf-life, especially in tropical climates.

“Dr. Sol Capote” heads the malaria research group at “Children’s Health International(CHI),” and he and his colleagues are now designing a large clinical trial to be coordinatedfrom “Jamkatnia” in West Africa. Dr. Capote is an experienced investigator, so he knowsthat substantial thought, effort, and experience must go into developing the sample-sizeanalysis and the rest of the statistical considerations.

The CHI study will use a randomized, double-blind design to compare usual care only(UCO) versus usual care plus a single dose of QCA. After reviewing all previous humanstudies of both DCA and QCA, the CHI team is convinced that a single dose of QCA isvery likely to be safe. Accordingly, after consulting with Jamkatnian health officials and abioethicist, they decide that two-thirds of the subjects should get QCA.

Page 254: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 241

10.3 p-Values, α, β and PowerThe primary efficacy analysis will yield a p-value that compares the mortality rates ofcontrol versus QCA. Smaller p-values indicate greater statistical separation between thetwo samples, but how that p-value is determined is an issue that is not critical tounderstanding the essential concepts in sample-size analysis. In this case, that p-value maycome from one of the many methods to compare two independent proportions, includingthe likelihood ratio chi-square test, as used here, or it may come from a logistic or hazardmodeling that includes co-predictors. Regardless of what test is used to get the p-value, if pis small enough (“significant”) and the QCA mortality rates are better, Dr. Capote willreport that the study supported the hypothesis that QCA reduces mortality in childrenwith severe malaria complicated with lactic acidosis. If the p-value is not small enough(“not significant”), then he will report that the data provided insufficient evidence tosupport the hypothesis.

10.3.1 Null and Non-Null Distributions of p-Values; Type I and Type II ErrorsDr. Capote’s quest here is to answer the following question: Does QCA decrease mortalityin children with severe malaria? While Mother Nature knows the correct answer, only if wewere able to gather an infinitely large, perfectly clean dataset could we figure this outourselves. Rather, we must design a study or, usually, a series of studies, that will yieldsample datasets that give us a solid chance of inferring what Mother Nature knows.Unfortunately, Lady Luck builds randomness into those sample datasets, and thus even thebest studies can deliver misleading answers.

Please study the top distribution in Figure 10.2. Here, there is no difference between thetwo groups’ mortality in the infinite dataset, so regardless of the sample size, all values for0 < p < 1 are equally likely. Accordingly, there is a 5% chance that p ≤ 0.05, or a 100α%chance that p ≤ α (in practice, these percentages are rarely exact because the data arediscrete or they fail to perfectly meet the test’s underlying mathematical assumptions). Ifthere is no true effect, but p ≤ α indicates otherwise, then this result triggers a Type Ierror, which is why α is called the Type I error rate. α should be chosen after somethought; it should not be automatically set at 0.05.

What if QCA has some true effect, good or bad? Then the non-null (non-central)distribution of the p-value will be skewed toward 0.0, as in the middle and bottom plots ofFigure 10.2. The middle one comes from presuming (1) true mortality rate of 0.28 for UCOand 0.21 for QCA, which is a 25% reduction in mortality; (2) 700 patients randomized toUCO versus 1400 to QCA, and (3) the p-value arises from testing whether the twomortality proportions differ (non-directional) using the likelihood ratio chi-square statistic.The bottom plot conforms to presuming that QCA cuts mortality by 33%.

Inferential power is the chance that p ≤ α when the null hypothesis is false, which is whyα could be called the “null power.” If there is some true effect, but p > α, then a Type IIerror is triggered. Consider the middle plot, which is based on a 25% reduction inmortality. Using the common Type I error rate, α = 0.05, the power is 0.68, so the Type IIerror rate is β = 0.32. By tolerating a higher α-level, we can increase power (decrease β).Here, using α = 0.20. the power is 0.87, so β = 0.13. If QCA is more effective (bottom plot,33% reduction in mortality), then the power rises to 0.91 with α = 0.05 and 0.98 withα = 0.20. Again, we will never know the true power, because Mother Nature will never tellus the true mortality rates in the two groups, and Lady Luck will always add some naturalrandomness into our outcome data.

10.3.2 Balancing Type I and II Error RatesRecall that Topol et al. (1997) advocated that the power should be around 90%, whichputs the Type II error rate around 10%. We generally agree, but stress that there should

Page 255: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

242 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 10.2 Distribution of the p-value under the null hypothesis and two non-null scenarios

α

β

α

β

α

β

α

β

be no standard power threshold that is accepted blindly as being satisfactory across allsituations. Why do so many people routinely perform power analyses using α = 0.05 and80% power (β = 0.20)? Rarely do they give it any thought.

Consider the middle plot in Figure 10.2. We could achieve a much better Type II errorrate of 13% if we are willing to accept a substantially greater Type I error rate of 20%.Investigators should seek to obtain α versus β values that are in line with the consequencesof making a Type I error versus a Type II error. In some cases, making a Type II error maybe far more costly than making a Type I error. In particular, in the early stages of theMarch of Science, making a Type I error may only extend the research to another stage.

Page 256: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 243

This is undesirable, of course, as are all mistaken inferences in science, but making a TypeII error may be far more problematic, because it may halt a line of research that wouldultimately be successful. So it might be justified to use α = 0.20 (maybe more) in order toreduce β as much as possible. Using such high α values is not standard, so investigatorsadopting this philosophy must be convincing in their argument.

10.4 A Classical Power AnalysisDr. Capote and his team plan their trial as follows.

Study Design

This trial follows the small (N = 62 + 62) DCA trial reported by Agbenyega et al. (2003).It will be double-blind, but instead of a 1:1 allocation, the team would like to considergiving one patient usual care only for every two patients that get QCA, where the QCA isgiven in a single infusion of 50 mg/kg.

SubjectsStudy patients will be less than 13 years old with severe malaria complicated by lacticacidosis. “Untreatable” cases (nearly certain to die) will be excluded. These terms willrequire operational definitions and the CHI team will formulate the otherinclusion/exclusion criteria and state them clearly in the protocol. They think it is feasibleto study up to 2100 subjects in a single malaria season using centers in Jamkatnia alone. Ifneeded, they can add more centers in neighboring “Gabrieland” and increase the total sizeto 2700. Drop-outs should not be a problem in this study, but all studies must consider thisand enlarge recruitment plans accordingly.

Primary Efficacy Outcome MeasureDeath before Day 10 after beginning therapy. Almost all subjects who survive to Day 10will have fully recovered. Time to death (i.e., survival analysis) is not a consideration.

Primary Analysis

To keep this story and example relatively simple, we will limit our attention to the basicrelative risk that associates treatment group (UCO vs. QCA) with death (no or yes). Forexample, if 10% died under QCA and 18% died under UCO, then the estimated relative riskwould be 0.10/0.18 = 0.55 in favor of QCA. p-values will be based on the likelihood ratiochi-square statistic for association in a 2 × 2 contingency table. The group’s biostatistician,“Dr. Phynd Gooden,” knows that the test of the treatment comparison could be made withgreater power through the use of a logistic regression model that includes baselinemeasurements such as a severity score or lactate levels, etc. (as was done in Holloway et al.,1995). In addition, this study will be completed in a single malaria season, so performinginterim analyses is not feasible. These issues are beyond the scope of this chapter.

10.4.1 Scenario for the Infinite DatasetA prospective sample-size analysis requires the investigators to characterize thehypothetical infinite dataset for their study. Too often, sample-size analysis reports fail toexplain the rationale undergirding the conjectures. If we explain little or nothing, reviewerswill question the depth of our thinking and planning, and thus the scientific integrity of ourproposal. We must be as thorough as possible and not apologize for having to make somesound guesstimates. All experienced reviewers have had to do this themselves.

Dr. Stacpoole’s N = 62 + 62 human study (Agbenyega et al., 2003) had eight deaths ineach group. This yields 95% confidence intervals of [5.7%, 23.9%] for the quinine-only

Page 257: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

244 Pharmaceutical Statistics Using SAS: A Practical Guide

mortality rate (using the EXACT statement’s BINOMIAL option in the FREQ procedure)and [0.40, 2.50] for the DCA relative risk (using the asymptotic RELRISK option in theOUTPUT statement in PROC FREQ). These wide intervals are of little help in specifyingthe scenario. However, CHI public health statistics and epidemiologic studies in theliterature indicate that about 19% of these patients die within ten days using quinine only.This figure will likely be lower for a clinical trial, because untreatable cases are beingexcluded and the general level of care could be much better than is typical. Finally, theHolloway et al. (1995) rat study obtained a DCA relative risk of 0.67 [95% CI: 0.44, 1.02],and the odds ratios adjusting for baseline covariates were somewhat more impressive, e.g.,OR= 0.46 (one-sided p = 0.021).

Given this information, the research team conjectures that the mortality rate is 12-15%for usual care. They agree that if QCA is effective, then it is reasonable to conjecture thatit will cut mortality 25-33% (relative risk of 0.67-0.75).

Topol et al. (1999) wrote about needing sufficient power to detect a clinically relevantdifference between the experimental and control therapies.” Some authors speak ofdesigning studies to detect the smallest effect that is clinically relevant. How do we definesuch things? Everyone would agree that mortality reductions of 25-33% are clinicallyrelevant. What about 15%? Even a 5% reduction in mortality would be considered veryclinically relevant in a disease that kills so many people annually, especially because asingle infusion of QCA is relatively inexpensive. Should the CHI team feel they must powerthis study to detect a 5% reduction in mortality? As we shall see, this is infeasible. It isusually best to ask: “What do we actually know at this point? What do we think ispossible? What scenarios are supportable? Will the reviewers agree with us?”

10.4.2 What Allocation Ratio? One-Sided or Two-Sided Test?Dr. Gooden is aware of the fact that the likelihood ratio chi-square test for two independentproportions can be more powerful when the sample sizes are unbalanced. Her first task isto assess how the planned 1:2 (UCO: QCA) allocation ratio affects the power. As shown inProgram 10.1, this is relatively easy to do in PROC POWER. Its syntax is literal enoughthat we will not explain it, but note particularly the GROUPWEIGHTS statement.

Table 10.1 displays results obtained using Program 10.1 and some simple furthercomputations. For this conjecture of 15% mortality versus 0.67 × 15% mortality, the mostefficient of these four designs is the 1:1 allocation ratio. It has a power of 0.930 or β = 0.070with Ntotal = 2100 (α = 0.05), and to get a 0.90 power requires Ntotal = 1870. Compared tothe 1:1 design, the 1:2 design has a 36% larger Type II error rate (“relative Type II riskratio”) at Ntotal = 2100 and requires 2064 subjects to achieve a 0.90 power. Thus, the 1:2design has a relative efficiency of 1870/2064 = 0.91 and requires about 10% more subjectsto achieve 0.90 power (relative inefficiency: 2064/1870 = 1.10). The relative inefficiencies forthe 2:3 and 1:3 designs are 1.03 and 1.29, respectively.

Table 10.1 Effect of the Allocation Ratio, NUCO : NQCA, on Power, β, and Sample Size for Two-Sidedα = 0.05 Assuming 15% Mortality with Usual Care and a Relative Risk of 0.67 in Favor of QCA

Allocation ratio (NUCO : NQCA)1:1 2:3 1:2 1:3

Power 0.930 0.923 0.905 0.855Ntotal = 2100 β 0.070 0.077 0.095 0.145

Relative Type II risk ratio 1.00 1.10 1.36 2.07

Ntotal 1870 1925 2064 2420Power = 0.90 Relative efficiency 1.00 0.97 0.91 0.77

Relative inefficiency 1.00 1.03 1.10 1.29

Page 258: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 245

Program 10.1 Compare allocation weights

* Powers at Ntotal=2100;proc power;

TwoSampleFreqGroupWeights = (1 1) (2 3) (1 2) (1 3) /* UCO:QCA */RefProportion = .15 /* Usual Care Only (UCO) mortality rate*/RelativeRisk = .67 /* QCA mortality vs. UCO mortality */alpha = .05sides = 1 2Ntotal = 2100test = lrchipower = .;

* Ntotal values for power = 0.90;proc power;

TwoSampleFreqGroupWeights = (1 1) (2 3) (1 2) (1 3)/* UCO:QCA */RefProportion = .15 /* Usual Care Only (UCO) mortality rate*/RelativeRisk = .67 /* QCA mortality vs. UCO mortality */alpha = .05sides = 1 2Ntotal = .test = lrchipower = .90;run;

Note that Dr. Gooden uses SIDES=1 2 in Program 10.1 to consider both one-sided andtwo-sided tests. Investigators and reviewers too often dogmatically call for two-sided testsonly because they believe using one-sided tests is not trustworthy. But being goodscientists, Dr. Capote’s team members think carefully about this issue. Some argue thatthe scientific question is simply whether QCA is efficacious versus whether it is notefficacious, where “not efficacious” means that QCA has no effect on mortality or itincreases mortality. This conforms to the one-sided test. For the design, scenario, andanalysis being considered here, the one-sided test requires 1683 subjects versus 2064 for thetwo-sided test, giving the two-sided test a relative inefficiency of 1.23. At N = 2100, theType II error rate for the one-sided test is β = 0.052, which is 45% less than the two-sidedrate of β = 0.095. On the other hand, other members argue that it is important to assesswhether QCA increases mortality. If it does, then the effective Type II error rate for theone-sided test is 1.00. This logic causes many to never view one-sided tests favorably underany circumstances. After considering these issues with Dr. Gooden, Dr. Capote decides totake the traditional approach and use a two-sided test.

For some endpoints, such as for rare adverse events or in trials involving rare diseases,the argument in favor of performing one-sided tests is often compelling. Suppose there issome fear that a potential new treatment for arthritis relief could increase the risk ofgastrointestinal bleeding in some pre-specified at-risk subpopulation, say, raising this froman incidence rate in the first 30 days from 8% to 24%, a relative risk of 3.0. A balancedtwo-arm trial with N = 450 + 450 subjects may be well-powered for testing efficacy(arthritis relief), but suppose the at-risk group is only 20% of the population beingsampled, so that only about N = 90 + 90 will be available for this planned sub-groupanalysis. Using α = 0.05, the likelihood ratio test for comparing two independentproportions will provide 0.847 power for the two-sided test and 0.910 power for theone-sided test. Thus, using a one-sided test cuts the Type II error rate from 0.153 to 0.090,a 41% reduction. Stated differently, using a two-sided test increases β by 70%. However, ifthis research aim is concerned only with detecting an increase in GI bleeding, why not use

Page 259: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

246 Pharmaceutical Statistics Using SAS: A Practical Guide

the statistical hypothesis—the one-sided version—that conforms to that aim? If using thetwo-sided test increases the Type II error rate by 70%, why is that more trustworthy?

For completeness, and because it takes so little time to do, Dr. Gooden also uses PROCPOWER to find the approximate optimal allocation ratio. After iterating the groupweights, she settles on using Program 10.2 to show that while the theoretical optimal isapproximately 0.485:0.515, the balanced (0.500:0.500) design has almost the same efficiency.

Program 10.2 Find optimal allocation weights

proc power;TwoSampleFreqGroupWeights = /* UCO : QCA */(.50 .50) (.49 .51) (.485 .515) (.48 .52) (.45 .55) (.33 .66)RefProportion = .15 /* Usual Care Only (UCO) mortality rate*/RelativeRisk = .67 /* QCA mortality vs. UCO mortality */alpha = .05sides = 2Ntotal = .test = LRchi /* likelihood ratio chi-square */power = .90nfractional;run;

Output from Program 10.2

Fractional Actual CeilingIndex Weight1 Weight2 N Total Power N Total

1 0.500 0.500 1868.510571 0.900 18692 0.490 0.510 1867.133078 0.900 18683 0.485 0.515 1867.002923 0.900 18684 0.480 0.520 1867.245653 0.900 18685 0.450 0.550 1876.616633 0.900 18776 0.330 0.660 2061.667869 0.900 2062

Should the study use the less efficient 1:2 design? After substantial debate within histeam, Dr. Capote decides that the non-statistical attributes of the 1:2 design give it morepractical power than the 1:1 design. First, nobody has safety concerns about giving a singledose of QCA. Second, Jamkatnian health officials and parents will prefer hearing that twoout of three subjects will be treated with something that could be life-saving for some.Third, the extra cost associated with a 10% increase in the sample size is not prohibitive.Given that this study’s set-up costs are high and the costs associated with data analysisand reporting are unaffected by the sample size, the total cost will increase by onlyabout 3%.

10.4.3 Obtaining and Tabling the PowersThe stage is now set to carry out and report the power analysis. Please examineProgram 10.3 together with Output 10.3, which contains the essential part of the results.In SAS 9.1, PROC POWER provides plain graphical displays of the results (not shownhere), but lacks corresponding table displays. As this chapter was going to press, ageneral-purpose SAS macro, %Powtable, was being developed to help meet this need; seethe book’s website. Here, Dr. Gooden uses the ODS OUTPUT command and theTABULATE procedure to create a basic table.

Page 260: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 247

Program 10.3 Power analysis for comparing mortality rates

options ls=80 nocenter FORMCHAR="|----|+|---+=|-/\<>*";

proc power;ODS output output=MortalityPowers;TwoSampleFreqGroupWeights = (1 2) /* 1 UCO : 2 QCA*/RefProportion = .12 .15 /* UCO mortality rate */RelativeRisk = .75 .67 /* QCA rate vs UCO rate*/alpha = .01 .05 .10sides = 2Ntotal = 2100 2700test = LRchi /* likelihood ratio chi-square */power = .;plot vary (panel by RefProportion RelativeRisk);

/* Avoid powers of 1.00 in table */data MortalityPowers;

set MortalityPowers;if power>0.999 then power999=0.999;else power999=power;

proc tabulate data=MortalityPowers format=4.3 order=data;format Alpha 4.3;class RefProportion RelativeRisk alpha NTotal;var Power999;tableRefProportion="Usual Care Mortality"

* RelativeRisk="QCA Relative Risk",alpha="Alpha"

* Ntotal="Total N"* Power999=""*mean=" "/rtspace=28;

run;

Output from Program 10.3

----------------------------------------------------------| | Alpha || |-----------------------------|| | .010 | .050 | .100 || |---------+---------+---------|| | Total N | Total N | Total N || |---------+---------+---------|| |2100|2700|2100|2700|2100|2700||--------------------------+----+----+----+----+----+----||Usual Care |QCA Relative | | | | | | ||Mortality |Risk | | | | | | ||------------+-------------| | | | | | ||0.12 |0.75 |.329|.437|.569|.677|.687|.780|| |-------------+----+----+----+----+----+----|| |0.67 |.622|.757|.823|.905|.893|.948||------------+-------------+----+----+----+----+----+----||0.15 |0.75 |.438|.566|.677|.783|.781|.864|| |-------------+----+----+----+----+----+----|| |0.67 |.757|.872|.905|.960|.948|.981|----------------------------------------------------------

Page 261: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

248 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 10.3 Plots for the mortality analysis showing how changing the reference proportion or the relative riskrate affects power.

αRel proportion=0.15, Rel risk=0.67

Total sample size2100 2700

rewo

P

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

Total sample size2100 2700

Rel proportion=0.12, Rel risk=0.75

Total sample size2100 2700

Rel proportion=0.15, Rel risk=0.75

Figure 10.3 juxtaposes three plots that were produced by using the same ODS outputdata set, MortalityPowers, with a SAS/GRAPH program not given here, but which isavailable at this book’s companion Website. This shows concretely how power increases forlarger UCO mortality rates or better (smaller) relative risks for QCA versus UCO.

Colleagues and reviewers should have little trouble understanding and interpreting thepowers displayed as per Output 10.3. If the goal is to have 0.90 power using α = 0.05, thenN = 2100 will suffice only under the most optimistic scenario considered: that is, if theusual care mortality rate is 15% and QCA reduces that risk 33%. N = 2700 seems to berequired to ensure adequate power over most of the conjecture space.

Tables like this are valuable for teaching central concepts in traditional hypothesistesting. We can see with concrete numbers how power is affected by various factors. Whilewe can set N and α, Mother Nature sets the mortality rate for usual care and the relativerisk associated with QCA efficacy.

Let us return to the phrase from Topol et al. (1997) that called for clinical trials to haveadequate power “to detect a clinically relevant difference between the experimental andcontrol therapies.” With respect to our malaria study, most people would agree that even atrue 5% reduction in mortality is clinically relevant and it would also be economicallyjustifiable to provide QCA treatment given its low cost and probable safety. But could wedetect such a small effect in this study? If QCA reduces mortality from 15% to0.95 × 15% = 14.25%, then the proposed design with N = 900 + 1800 has only 0.08 power(two-sided α = 0.05). In fact, under this 1:2 design and scenario, it will require almost104,700 patients to provide 0.90 power. This exemplifies why confirmatory trials (PhaseIII) are usually designed to detect plausible outcome differences that are considerablylarger than “clinically relevant.” The plausibility of a given scenario is based on biologicalprinciples and from data gathered in previous relevant studies of all kinds and qualities. Byordinary human nature, investigators and statisticians tend to be overly optimistic inguesstimating what Mother Nature might have set forth, and this causes our studies to beunderpowered. This problem is particularly relevant when new therapies are tested againstexisting therapies that might be quite effective already. It is often the case that potentiallysmall but important improvements in therapies can be reliably assessed only in very largetrials. Biostatisticians are unwelcome and even sometimes disdained when they bring thisnews, but they did not make the Fundamental Laws of Chance—they are only chargedwith policing and adjudicating them.

Page 262: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 249

10.5 Beyond α and β: Crucial Type I and Type II Error RatesAre α and β (or power= 1 − β) the only good ways to quantify the risk of making Type Iand Type II errors? While they may be the classical rates to consider and report, they failto directly address two fundamental questions:

• If the trial yields traditional statistical significance (p ≤ α), what is the chance this willbe an incorrect inference?

• If the trial does not yield traditional statistical significance (p > α), what is the chancethis will be an incorrect inference?

To answer these in some reasonable way, we need to go beyond using just α and β.

10.5.1 A Little Quiz: Which Study Provides the Strongest Evidence?Table 10.2 summarizes outcomes from three possible QCA trials. Which study has thestrongest evidence that QCA is effective? Studies #1 and #2 have N = 150 + 300 subjects,whereas #3 has N = 700 + 1400 subjects. Studies #1 and #3 have identical 0.79 estimatesof relative risk, but with p = 0.36, Study #1 does not adequately support QCA efficacy.Choosing between Studies #2 and #3 is harder. They have the same p-value, so manypeople would argue that they have the same inferential support. If so, then #2 is thestrongest result, because its relative risk of 0.57 is substantially lower than the relative riskof 0.79 found in Study #3. However, Study #3 has nearly five times the sample size, so ithas greater power. How should that affect our assessment?

Table 10.2 Which Study Has the Strongest Evidence That QCA is Effective?

Deaths/N Mortality Relative riskStudy UCO QCA UCO QCA RR [95% CI]

LR testp-value

#1 21/150 33/300 14.0% 11.0% 0.79 [0.47, 1.31] 0.36#2 21/150 24/300 14.0% 8.0% 0.57 [0.33, 0.992] 0.05#3 98/700 154/1400 14.0% 11.0% 0.79 [0.62, 0.995] 0.05

10.5.2 To Answer the Quiz: Compare the Studies’ Crucial Error RatesSuppose that Mother Nature has set the true usual care mortality rate at 0.15 and theQCA relative risk at 0.67, the most powerful scenario we considered above. We havealready seen (Figure 10.3, Output 10.3) that with N = 700 + 1400 subjects and usingα = 0.05 (two-sided), the power is 90%. With 150 subjects getting usual care and 300getting QCA, the power is only about 33%.

Now, in addition, suppose that Dr. Capote and his team are quite optimistic that QCAis effective. This does not mean they have lost their ordinary scientific skepticism andalready believe that QCA is effective. Consider another Feynman-ism (1999, P. 200):

The thing that’s unusual about good scientists is that they’re not so sure of themselves asothers usually are. They can live with steady doubt, think “maybe it’s so” and act on that, allthe time knowing it’s only “maybe.”

Dr. Capote’s team understands that even for the most promising experimentaltreatments, the clear majority fail to work when tested extensively. In fact, Lee and Zelen(2000) estimated that among 87 trials completed and reported out by the EasternCooperative Oncology Group at Harvard from 1980-1995, only about 30% seem to havebeen testing therapies that had some clinical efficacy.

Page 263: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

250 Pharmaceutical Statistics Using SAS: A Practical Guide

Let us suppose that Dr. Capote’s team conducted 1000 independent trials looking forsignificant treatment effects, but Mother Nature had set things up so that 700 effects wereactually null. What would we expect to happen if Dr. Capote ran all 1000 trials at averagepowers of 33%? 90%? Table 10.3 presents some straightforward computations thatillustrate what we call the crucial Type I and Type II error rates. With 700 null tests, wewould expect to get 35 (5%) Type I errors (false positives). From the 300 non-nullhypotheses tested with 33% power, we would expect to get 99 true positives. Thus, each“significant” test (p ≤ 0.05) has an α∗ = 35/134 = 0.26 chance of being misleading. Notehow much larger this is than α = 0.05. Some people (including authors of successfulstatistics books) confuse α and α∗, and hence they also misinterpret what p-values are. Ap-value of 0.032 does not imply that there is a 0.032 chance that the null hypothesis is true.

Table 10.3 Expected Results for 1000 Tests Run at α = 0.05. Note: The true hypothesis is null in 700 tests.For the 300 non-null tests, the average power is 33% or 90%.

Result of hypothesis testp ≤ 0.05 (“significant”) p > 0.05 (“not significant”)

33% average power

700 true null 5% of 700 = 35 95% of 700 = 665300 true non-null 33% of 300 = 99 67% of 300 = 201

Crucial Type I error rate: Crucial Type II error rate:α∗ = 35/134 = 0.26 β∗ = 201/866 = 0.23

90% average power

700 true null 5% of 700 = 35 95% of 700 = 665300 true non-null 90% of 300 = 270 10% of 300 = 30

Crucial Type I error rate: Crucial Type II error rate:α∗ = 35/305 = 0.11 β∗ = 30/695 = 0.04

The crucial Type II error rate, β∗, is defined similarly. With 33% power, we wouldexpect to get 201 Type II errors (false negatives) to go with 665 true negatives; thus β∗ =210/866 = 0.23. Note that this is not equal to β = 0.67.

10.5.3 Greater Power Reduces Both Types of Crucial Error RatesA key point illustrated in Table 10.3 is that greater power reduces both types of crucial errorrates. In other words, statistical inferences are generally more trustworthy when theunderlying power is greater. Let us return to Table 10.2. Again, which study has thestrongest evidence that QCA is effective? Even under our most powerful scenario, ap ≤ 0.05 result has a 0.26 chance of being misleading when using N = 150 + 300, as perStudy #2. This falls to 0.11 using N = 700 + 1400 (Study #3). Both studies may haveyielded p = 0.05, but they do not provide the same level of support for inferring that QCAis effective. Study #3 provides the strongest evidence that QCA has some degree ofefficacy. This concept is poorly understood throughout all of science.

10.5.4 The March of Science and Sample-Size AnalysisConsistent with Lee and Zelen (2000), we think that investigators designing clinical trialsare well served by considering α∗ and β∗. (Note that Lee and Zelen’s definition is reversedfrom ours in that our α∗ and β∗ correspond to their β∗ and α∗, respectively.) Ioannidis(2005b) used the same logic in arguing “why most published research findings are false.”Wacholder et al. (2004) described the same methodology to more carefully infer whether agenetic variant is really associated with a disease. Their “false positive report probability”

Page 264: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 251

is identical to α∗. Also, readers familiar with accuracy statistics for medical tests will seethat 1 − α and β are isomorphic to the specificity and sensitivity of the diagnostic methodand 1 − α∗ and 1 − β∗ are isomorphic with the positive and negative predictive values.

Formally, let γ be the probability that the null hypothesis is false. We like to think of γas measuring where the state of knowledge currently is in terms of confirming the non-nullhypothesis; in short, its location along its March of Science (Figure 10.1). Thus, for novelresearch hypotheses, γ will be nearer to 0. For mature hypotheses that are ready for solidconfirmation with say, a large Phase III trial, γ will be markedly greater than 0. We mightregard γ = 0.5 as scientific equipoise, saying that the hypothesis is halfway along its pathto absolute confirmation in that we consider the null and non-null hypothesis as equallyviable. Lee and Zelen’s (2000) calculations put γ around 0.3 for Phase III trials coordinatedin the Eastern Cooperative Oncology Group.

Given γ, α and some β set by some particular design, sample size, and non-null scenario,we can apply Bayes’ Theorem to get

α∗ = Prob[H0 true | p ≤ α] =α(1 − γ)

α(1 − γ) + (1 − β)γ

and

β∗ = Prob[H0 false | p > α] =βγ

βγ + (1 − α)(1 − γ).

To be precise, “H0 false” really means “H0 false, as conjectured in some specific manner.”For the example illustrated first in Table 10.3, we have γ = 0.30, α = 0.05 and β = 0.67,thus

α∗ =(0.05)(1 − 0.30)

(0.05)(1 − 0.30) + (1 − 0.67)(0.30)= 0.261

and

β∗ =(0.67)(0.30)

(0.67)(0.30) + (1 − 0.05)(1 − 0.30)= 0.232.

In Bayesian terminology, γ = 0.3 is the prior probability that QCA is effective, and1 − α∗ = 0.739 is the posterior probability given that p ≤ α. However, nothing here involvesBayesian data analysis methods, which have much to offer in clinical research, but are notgermane to this chapter. Some people are bothered by the subjectivity involved inspecifying prior probabilities like γ, but we counter by pointing out that there are manyother subjectivities involved in sample-size analysis for study planning, especially theconjectures made in defining the infinite dataset. Indeed, we find that most investigatorsare comfortable specifying γ, at least with a range of values, and that computing various α∗

and β∗ values of interest gives them much better insights into the true inferential strengthof their proposed (frequentist) analyses.

10.6 Research Question 1, Continued: Crucial Error Rates forMortality AnalysisIn developing the statistical considerations for the QCA/malaria trial, Dr. Goodenunderstands the value in assessing its crucial Type I and Type II error rates, and shepresses her CHI colleagues to complete the exercise faithfully. As mentioned before, theyare optimistic that QCA is effective, but to compute crucial error rates, they must nowquantify that optimism by setting γ. Initial discussions place γ near 0.75, but the 0.30value reported by Lee and Zelen (2000) tempers their thinking substantially. They settle onγ = 0.50. Dr. Gooden will also use γ = 0.30.

Page 265: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

252 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 10.4 Compute crucial error rates for mortality endpoint

options ls=80 nocenter FORMCHAR="|----|+|---+=|-/\<>*";

proc power;ODS output output=MortalityPowers;TwoSampleFreqGroupWeights = (1 2) /* 1 UCO for every 2 QCA */RefProportion = .12 .15 /* UCO mortality rate */RelativeRisk = .75 .67 /* QCA rate vs UCO rate */alpha = .01 .05sides = 2Ntotal = 2700test = LRchi /* likelihood ratio chi-square */power = .;

plot key=OnCurves;run;

* Call %CrucialRates macro, given in Appendix B of this chapter;%CrucialRates( PriorPNullFalse = .30 .50,

Powers = MortalityPowers,CrucialErrRates = MortalityCrucRates )

proc tabulate data=MortalityCrucRates format=4.3 order=data;title3 "Crucial Error Rates for QCA Malaria Trial";format alpha 4.3;class RefProportion RelativeRisk alpha gamma TypeError NTotal;var CrucialRate;tableNtotal="Total N: ",RefProportion="Usual Care Mortality"

* RelativeRisk="QCA Relative Risk",alpha="Alpha"

* gamma="PriorP[Null False]"* TypeError="Crucial Error Rate"* CrucialRate=""*mean=" "

/ rtspace = 26;run;

Program 10.4 gives the code to handle this. First, a more focused version ofProgram 10.3 computes the powers. Second, a macro called %CrucialRates (given inAppendix B and available on the book’s web site) converts the PROC POWER results intocrucial Type I and Type II error rates. Finally, PROC TABULATE organizes these crucialrates effectively.

Output 10.4 shows that the most optimistic case considered here presumes that themortality rate is 0.15 under usual care, and it takes QCA to have a prior probability ofγ = 0.50 of being effective, where “effective” is a QCA relative risk of 0.67. If so, then byusing α = 0.05, the crucial Type I and Type II error rates are α∗ = 0.050 and β∗ = 0.040,respectively, which seem very good. However, for α = 0.01, β∗ rises to 0.115. Now considerthe most pessimistic case. If γ = 0.30 and the non-null scenario has a mortality rate of 0.12under usual care and a QCA relative risk is 0.75, then using α = 0.05 gives α∗ = 0.147 andβ∗ = 0.127. The team from Children’s Health International decides that they can toleratethese values and, thus, planning continues around N = 2700.

Page 266: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 253

After going through this process, Dr. Capote remarks that if all clinical trial protocolswere vetted in this manner, a great many of them would show crucial Type I and Type IIerror rates that would severely temper any inferences that can be made. This is true.

Output from Program 10.4

Total N: 2700------------------------------------------------------------------| | Alpha || |---------------------------------------|| | .010 | .050 || |-------------------+-------------------|| |PriorP[Null False] |PriorP[Null False] || |-------------------+-------------------|| | 0.3 | 0.5 | 0.3 | 0.5 || |---------+---------+---------+---------|| | Crucial | Crucial | Crucial | Crucial || | Error | Error | Error | Error || | Rate | Rate | Rate | Rate || |---------+---------+---------+---------|| |Type|Type|Type|Type|Type|Type|Type|Type|| | I | II | I | II | I | II | I | II ||------------------------+----+----+----+----+----+----+----+----||Usual Care |QCA Relative| | | | | | | | ||Mortality |Risk | | | | | | | | ||-----------+------------| | | | | | | | ||0.12 |0.75 |.051|.196|.022|.362|.147|.127|.069|.254|| |------------+----+----+----+----+----+----+----+----|| |0.67 |.030|.095|.013|.197|.114|.041|.052|.091||-----------+------------+----+----+----+----+----+----+----+----||0.15 |0.75 |.040|.158|.017|.305|.130|.089|.060|.186|| |------------+----+----+----+----+----+----+----+----|| |0.67 |.026|.053|.011|.115|.108|.018|.050|.040|------------------------------------------------------------------

10.7 Research Question 2: Does “QCA” Affect the“Elysemine : Elysemate” Ratios (EER)?This section expands Dr. Capote’s planning to consider a test that compares the UCO andQCA arms with respect to a continuous outcome, adjusted for baseline covariates. PROCGLMPOWER is used to perform the calculations.

10.7.1 Rationale Behind the Research QuestionNow the team turns to investigating potential adverse effects.

A descriptive analysis being completed in Jamkatnia has compared 34 children who havesevere malaria with 42 healthy children on some 27 measures related to metabolicfunctioning, including two amino acids, “elysemine” and “elysemate” (both fictitious).Elysemine is synthesized by the body from elysemate, which is abundant in food grains andmeat. Phagocytes (a type of white blood cell) need elysemine to fight infection. Lowplasma elysemine levels have been shown to be an incremental risk factor for death incritically ill adults and children, especially in very premature infants. Thus, a suppressedelysemine:elysemate ratio (EER) seems to be associated with a weakened immune system.In addition, plasma elysemine concentrations fall, and plasma elysemate concentrationsrise, in response to extended periods of physical exertion, such as marathon running. Of

Page 267: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

254 Pharmaceutical Statistics Using SAS: A Practical Guide

course, typical marathon runners have no problem rapidly converting elysemate toelysemine and their EERs rebound within two hours.

This Jamkatnian study is of keen interest because the children with malaria had amedian EER of 2.00 (inter-95% range: 1.10-3.04) compared to 2.27 (inter-95% range:1.50-3.28) for the healthy children (p = 0.01, two-tailed median test). The researchers nowrationalize that children with severe malaria may show reduced EERs, because the parasiteattacks red blood cells and this reduces blood oxygen levels. Given that so many measureswere analyzed in an exploratory manner, this p = 0.01 result is supportive, but notconfirmatory. Nevertheless, it stirs great attention.

Related to this was a study of seven healthy adult human volunteers who were given asingle standard dose of QCA and monitored intensively for 24 hours in a General ClinicalResearch Center. The data are summarized in Table 10.4. By four hours post infusion,their EERs fell by a geometric average of 14.9% (p = 0.012; 95% CI: 4.9-23.8% reductionvia one-sample, two-sided t test comparing log(EER) values measured pre and post). Inthat the EER may already be suppressed in these diseased children, any further reductioncaused by QCA would be considered harmful. On the other hand, EERs could rebound(rise) more quickly as QCA reduces lactic acid levels and thus helps restore metabolicnormality. Accordingly, now the research question is “Does QCA increase or decreaseelysemine:elysemate ratios in children with severe malaria complicated by lactic acidosis?”

Table 10.4 Elysemine and Elysemate Levels from Seven Healthy Adults Given QCA

Baseline 4 Hours After QCASubject E’mine0 E’mate0 EER0 E’mine4 E’mate4 EER4 EER4/EER0

1 288 143 2.01 260 167 1.56 0.772 357 163 2.19 302 135 2.24 1.023 285 122 2.34 246 129 1.91 0.824 349 143 2.44 317 157 2.02 0.835 332 127 2.61 285 152 1.88 0.726 329 119 2.76 294 114 2.58 0.937 389 114 3.41 365 118 3.09 0.91

Geometricmean 331 132 2.51 293 138 2.13 0.85

Upper 95%limit 367 149 2.94 331 158 2.63 0.95

Lower 95%limit 298 117 2.14 260 120 1.73 0.76

Review of Study Design and SubjectsTo reiterate, this double-blinded trial will randomize 900 subjects to receive usual care only(UCO) and 1800 to receive a single infusion of 50 mg/kg QCA. Study patients will be lessthan 13 years old diagnosed with severe malaria complicated by lactic acidosis.

Continuous Outcome Measure and Baseline Covariates

Our focus here is on the elysemine:elysemate ratio measured four hours post-infusion(EER4). The three primary covariates being considered are the baseline (five minutes priorto QCA infusion) measures of log EER0, plasma lactate level, and log parasitemia, thepercentage of red blood cells infected.

Page 268: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 255

It should be mentioned that elysemine and elysemate assays are expensive to conduct,costing about US$60 for each time, and thus costing US$120 for each subject.

Planned AnalysisRatio measurements like EER are usually best handled after being log transformed; forease of understanding we shall use log2(EER4), so that a 1.0 log discrepancy between twovalues equates to having one value twice that of the other.

Scenarios for the Infinite Datasets

Figure 10.4 Scenario for EER4 distributions of the Usual Care Only and QCA arms. Note: The medians, as wellas the geometric means, are 2.0 and 1.8, and the common inter-95% relative spread is 3.16/1.26 = 2.85/1.14 =2.5.

���

���

���

��

!�� ���"!�� ����#����

$���� ����%

�σ&'(

����

���)

���)

σ)*(

���

���

��)

���

����$!�� ���+!�� ���%

, ���-�.���

, ���-�/�0-1

��*�

��*'

���

Based on the Jamkatnian study reviewed above, the investigators speculate that themedian EER4 for the Usual Care Only arm is 2.0. See Figure 10.4. Two scenarios for theQCA arm are considered, a 10% decrease in EER4 (2.0 versus 1.8; as per Figure 10.4) and a15% decrease (2.0 versus 1.7). Assuming that log2(EER4) has a Normal distribution, EER4medians of 2.0 versus 1.8 (or 1.7) become log2(EER4) means of 1.00 versus 0.85 (or 0.77).

Making conjectures for the spread is a knotty problem, and the values chosen havecritical influence on the sample-size analysis. Dr. Gooden usually takes a pragmaticapproach based on the fact that, for a Normal distribution, the inter-95% range spansabout four standard deviations. Thus, when the outcome variable is Normal, it is sufficientto estimate or guesstimate the range of the middle 95% of the infinite dataset for a groupand divide by four to set the scenario for the standard deviation.

Here, Dr. Gooden takes log(EER4) to be Normal, i.e. EER4 is logNormal, so the processis a bit more complex. Let EER40.025 and EER40.975 be the 2.5% and 97.5% quantiles of adistribution of EER4 values. With respect to log2(EER4) values, the approximate standarddeviation is

σ =log2(EER40.975) − log2(EER40.025)

4=

log2(EER40.975/EER0.025)4

.

Define RS95 = EER40.975/EER40.025 to be the inter-95% relative spread of EER4. Forthe Jamkatnian study, these were 3.04/1.10 = 2.76 and 3.28/1.50 = 2.18. To be

Page 269: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

256 Pharmaceutical Statistics Using SAS: A Practical Guide

conservative, Dr. Capote sets RS95 to be either 2.5 (as per Figure 10.4) or 3.0. Both armsare assumed to have the same relative spread. These give values for σ of log2(2.5)/4 = 0.33and log2(3.0)/4 = 0.40.

Now Dr. Gooden needs to have the team decide how strongly the three baselinecovariates are correlated to log2(EER4). Technically, this correlation is the partial multiplecorrelation, R, of X1 = log2(EER0), X2 = plasma lactate, and X3 = log2(parasitemia) withY = log2(EER4), controlling for treatment group, but this terminology is not likely to bewell understood by the CHI team. Is there any existing data on this? Not for childreninfected with malaria. So, Gooden asks Dr. Capote’s group to imagine that some baselineindex is computed by taking a linear combination of the three covariates(b1X1 + b2X2 + b3X3) in such a way that this index is maximally correlated withlog2(EER4) within the two treatment groups. Dr. Gooden needs to know what R might bein the infinite dataset, but she does not simply ask them this directly, because fewinvestigators have good understandings about what a given correlation value, say ρ = 0.30,conveys. Instead, she shows them a version of Figure 10.5 that has the values of thecorrelations covered from view.

The strongest correlation is most likely to be between log2(EER0) and log2(EER4). Theteam agrees and suspects that this is at least ρ = 0.20, even if the malaria and thetreatments have a substantial impact on the metabolic pathways affecting EER. Usingplasma lactate and parasitemia to also predict log2(EER4) can only increase R. Looking atthe scatterplots in Figure 10.5, the team agrees that R is, conservatively, between 0.20 and0.50.

Figure 10.5 Scatterplots showing eight degrees of correlation. The order of presentation is unsystematic to aidin eliciting more careful conjectures.

Finally, Dr. Capote wants a minimal risk of committing a Type I or Type II error forthis question, so he would like to keep both α and β levels below 0.05. We will investigatethe crucial error rates, α∗ and β∗, later.

Page 270: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 257

Classical Power AnalysisIn order for SAS to compute the powers for this problem, two programming steps arenecessary. First, Program 10.5 creates an “exemplary” dataset that conforms to theconjectured infinite dataset.

Program 10.5 Build and print an exemplary dataset

data EER;group = "UCO";

CellWgt = 1;meanlog2EER_a = log2(2.0);meanlog2EER_b = log2(2.0);output;

group = "QCA";CellWgt = 2;meanlog2EER_a = log2(1.8);meanlog2EER_b = log2(1.7);output;

run;

proc print data=EER;run;

The PROC PRINT output shows that there are only two exemplary cases in thedataset, one to specify the UCO group and the other to specify the QCA group.

Output from Program 10.5

Obs group CellWgt meanlog2EER_a meanlog2EER_b1 UCO 1 1.00000 1.000002 QCA 2 0.84800 0.76553

Secondly, Program 10.6 analyzes the exemplary dataset using PROC GLMPOWER.

Program 10.6 Use PROC GLMPOWER to see range of Ntotal values

proc GLMpower data=EER;ODS output output=EER_Ntotals;class group;model meanlog2EER_a meanlog2EER_b = group;weight CellWgt;power

StdDev = 0.33 0.40 /* log2(2.5)/4 and log2(3.0)/4 */Ncovariates = 3CorrXY = .2 .35 .50alpha = .01 .05power = 0.95 0.99Ntotal = .;

run;

Lastly, Program 10.7 summarizes the Ntotal values in a basic, but effective manner(Output 10.7). Again, more sophisticated methods are possible.

Page 271: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

258 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 10.7 Table the Ntotal values

* Augment GLMPOWER output to facilitate tabling ;data EER_Ntotals; set EER_Ntotals;if dependent = "meanlog2EER_a" then EEratio = "2.0 vs 1.8";if dependent = "meanlog2EER_b" then EEratio = "2.0 vs 1.7";;if UnadjStdDev = 0.33 then RelSpread95 = 2.5;if UnadjStdDev = 0.40 then RelSpread95 = 3.0;run;

proc tabulate data=EER_Ntotals format=5.0 order=data;format Alpha 4.3 RelSpread95 3.1;class EEratio alpha RelSpread95 CorrXY NominalPower;var Ntotal;tableEEratio="EE Ratios: "

* alpha="Alpha"* NominalPower="Power",

RelSpread95="95% Relative Spread"* CorrXY="Partial R for Covariates"* Ntotal=""*mean=" "

/rtspace=35;run;

Output from Program 10.7

-----------------------------------------------------------------------| | 95% Relative Spread || |-----------------------------------|| | 2.5 | 3.0 || |-----------------+-----------------|| | Partial R for | Partial R for || | Covariates | Covariates || |-----------------+-----------------|| |0.20 |0.35 |0.50 |0.20 |0.35 |0.50 ||---------------------------------+-----+-----+-----+-----+-----+-----||EE Ratios:|Alpha |Power | | | | | | ||----------+----------+-----------| | | | | | ||2.0 vs 1.8|.010 |0.95 | 369| 336| 288| 537| 492| 420|| | |-----------+-----+-----+-----+-----+-----+-----|| | |0.99 | 495| 453| 387| 723| 663| 567|| |----------+-----------+-----+-----+-----+-----+-----+-----|| |.050 |0.95 | 267| 246| 210| 393| 360| 306|| | |-----------+-----+-----+-----+-----+-----+-----|| | |0.99 | 378| 345| 297| 552| 507| 432||----------+----------+-----------+-----+-----+-----+-----+-----+-----||2.0 vs 1.7|.010 |0.95 | 156| 144| 123| 228| 210| 180|| | |-----------+-----+-----+-----+-----+-----+-----|| | |0.99 | 210| 192| 165| 306| 282| 240|| |----------+-----------+-----+-----+-----+-----+-----+-----|| |.050 |0.95 | 114| 105| 90| 168| 153| 132|| | |-----------+-----+-----+-----+-----+-----+-----|| | |0.99 | 162| 147| 126| 234| 216| 183|-----------------------------------------------------------------------

Page 272: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 259

Upon scanning the results in Output 10.7, Drs. Capote and Gooden decide thatNtotal = 100 + 200 may be minimally sufficient, and Gooden focuses on this by usingProgram 10.8.

Program 10.8 Compute and table powers at Ntotal = 300 for EER4 outcome

proc GLMpower data=EER;ODS output output=EER_powers;class group;model meanlog2EER_a meanlog2EER_b = group;weight CellWgt;power

StdDev = 0.33 0.40 /* log2(2.5)/4 and log2(3.0)/4 */Ncovariates = 3CorrXY = .2 .35 .5alpha = .01 .05Ntotal = 300power = .;

run;

* Augment GLMPOWER output to facilitate tabling ;data EER_powers; set EER_powers;if dependent = "meanlog2EER_a" then EEratio = "2.0 vs 1.8";if dependent = "meanlog2EER_b" then EEratio = "2.0 vs 1.7";;if UnadjStdDev = 0.33 then RelSpread95 = 2.5;if UnadjStdDev = 0.40 then RelSpread95 = 3.0;if power > .999 then power999 = .999;

else power999 = power;run;

proc tabulate data=EER_powers format=4.3 order=data;format Alpha 4.3 RelSpread95 3.1;class EEratio alpha RelSpread95 CorrXY Ntotal;

var power999;tableNtotal="Total Sample Size: ",EEratio="EE Ratios: "* alpha="Alpha",

RelSpread95="95% Relative Spread"* CorrXY="Partial R for Covariates"* power999=""*mean=" "

/rtspace=35;run;

Output 10.8 shows that only in the most pessimistic scenario does the power drop alittle below 0.90 using Ntotal = 300 and α = 0.05, and the mid-range scenarios even havesubstantial power at α = 0.01. Furthermore, with Ntotal = 300, the assay costs associatedwith this aim will run about 300 × US$120 = US$36000, which is deemed practical. TheCHI team still wants to assess the crucial Type I and Type II error rates.

Page 273: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

260 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 10.8

Total Sample Size: 300-----------------------------------------------------------------| | 95% Relative Spread || |-----------------------------|| | 2.5 | 3.0 || |--------------+--------------|| |Partial R for |Partial R for || | Covariates | Covariates || |--------------+--------------|| |0.20|0.35|0.50|0.20|0.35|0.50||---------------------------------+----+----+----+----+----+----||EE Ratios: |Alpha | | | | | | ||----------------+----------------| | | | | | ||2.0 vs 1.8 |.010 |.893|.922|.959|.717|.764|.838|| |----------------+----+----+----+----+----+----|| |.050 |.969|.979|.991|.884|.910|.946||----------------+----------------+----+----+----+----+----+----||2.0 vs 1.7 |.010 |.999|.999|.999|.989|.994|.998|| |----------------+----+----+----+----+----+----|| |.050 |.999|.999|.999|.998|.999|.999|-----------------------------------------------------------------

10.7.2 Crucial Type I and Type II Error RatesBased on the current state of knowledge reviewed above, Dr. Capote’s team believes thatwhile this hypothesis is important to investigate seriously, there is only a 20-30% chancethat QCA affects EER. Accordingly, Dr. Gooden uses Program 10.9 to convert the resultsgiven in Output 10.8 to the crucial error rates.

Program 10.9 Compute and table crucial error rates for EER4 outcome

%CrucialRates ( PriorPNullFalse= .20 .30,Powers = EER_powers,CrucialErrRates = EERCrucRates )

proc tabulate data=EERCrucRates format=4.3 order=data;title3 "Crucial Error Rates for EER Outcome";format Alpha 4.3 RelSpread95 3.1;class TypeError gamma EEratio alpha RelSpread95 CorrXY Ntotal;var CrucialRate;tableNtotal="Total N: ",EEratio="EE Ratios: "

* RelSpread95="95% Relative Spread"* CorrXY="Partial R for Covariates",

alpha="Alpha"* gamma="PriorP[Null False]"* TypeError="Crucial Error Rate"* CrucialRate=""*mean=" "

/ rtspace = 32;run;

Page 274: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 261

Output from Program 10.9

Total N: 300------------------------------------------------------------------------| | Alpha || |---------------------------------------|| | .010 | .050 || |-------------------+-------------------|| |PriorP[Null False] |PriorP[Null False] || |-------------------+-------------------|| | 0.2 | 0.3 | 0.2 | 0.3 || |---------+---------+---------+---------|| | Crucial | Crucial | Crucial | Crucial || | Error | Error | Error | Error || | Rate | Rate | Rate | Rate || |---------+---------+---------+---------|| |Type|Type|Type|Type|Type|Type|Type|Type|| | I | II | I | II | I | II | I | II ||------------------------------+----+----+----+----+----+----+----+----||EE |95% |Partial R | | | | | | | | ||Ratios: |Relative |for | | | | | | | | ||---------|Spread |Covariates| | | | | | | | ||2.0 vs |---------+----------| | | | | | | | ||1.8 |2.5 |0.20 |.043|.026|.025|.044|.171|.008|.107|.014|| | |----------+----+----+----+----+----+----+----+----|| | |0.35 |.042|.019|.025|.033|.170|.005|.106|.009|| | |----------+----+----+----+----+----+----+----+----|| | |0.50 |.040|.010|.024|.017|.168|.002|.105|.004|| |---------+----------+----+----+----+----+----+----+----+----|| |3.0 |0.20 |.053|.067|.032|.109|.184|.030|.117|.050|| | |----------+----+----+----+----+----+----+----+----|| | |0.35 |.050|.056|.030|.093|.180|.023|.114|.039|| | |----------+----+----+----+----+----+----+----+----|| | |0.50 |.046|.039|.027|.065|.174|.014|.110|.024||---------+---------+----------+----+----+----+----+----+----+----+----||2.0 vs |2.5 |0.20 |.038|.000|.023|.000|.167|.000|.104|.000||1.7 | |----------+----+----+----+----+----+----+----+----|| | |0.35 |.038|.000|.023|.000|.167|.000|.104|.000|| | |----------+----+----+----+----+----+----+----+----|| | |0.50 |.038|.000|.023|.000|.167|.000|.104|.000|| |---------+----------+----+----+----+----+----+----+----+----|| |3.0 |0.20 |.039|.003|.023|.005|.167|.000|.105|.001|| | |----------+----+----+----+----+----+----+----+----|| | |0.35 |.039|.002|.023|.003|.167|.000|.105|.000|| | |----------+----+----+----+----+----+----+----+----|| | |0.50 |.039|.000|.023|.001|.167|.000|.104|.000|------------------------------------------------------------------------

Dr. Capote likes what he sees here using α = 0.01, because almost all the α∗ and β∗

values are less than 0.05. The CHI team decides to use α = 0.01 and Ntotal = 100 + 200subjects for the EER component of this trial.

10.7.3 Using Baseline Covariates in Randomized StudiesWhat are the consequences of failing to use helpful baseline covariates when comparingadjusted group means in randomized designs? What are the consequences of usingworthless baseline covariates—those that have no value whatsoever in predicting the

Page 275: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

262 Pharmaceutical Statistics Using SAS: A Practical Guide

outcome (Y )? Researchers face this question because each additional covariate requiresanother parameter to be estimated, and this decreases by 1 the degrees of freedom for errorfor the F test of the group differences.

The question is easily addressed, and the answer surprises many. The power valuesdisplayed in Table 10.5 were obtained by modifying the PROC GLMPOWER code inProgram 10.8. Here, we limit our focus to the case with EER medians of 2.0 versus 1.8, a95% relative spread of 2.5, Ntotal = 300, and α = 0.01. On the other hand, we considerseveral more values for R (SAS Code: CorrXY = 0 .20 .35 .50 .70) and three possiblevalues for the number of covariates (SAS Code: Ncovariates = 0 3 50).

Table 10.5 Powers Obtained Using or Not Using Baseline Covariates in Randomized Studies

Multiple partial correlation (R)Number ofcovariates used 0.00 0.20 0.35 0.50 0.70

0 .878 .878 .878 .878 .8783 .878 .893 .922 .959 .996

50 .877 .892 .921 .959 .996

The point here is obvious. In a randomized design, there is virtually no cost associatedwith using worthless baseline covariates, because they are uncorrelated with the groupassignment. The only cost is that the nominal null F distributions change, but in this case,the 0.01 critical values for F(1, 298) and F(1, 248) are 6.72 and 6.74, respectively, whichare virtually equal. On the other hand, there is a high cost to be paid by not using baselinecovariates that have some value in predicting the outcome. This concept holds for bothcontinuous and categorical outcomes.

10.8 Crucial Error Rates When the Null Hypothesis Is Likely to Be TrueSuppose “Dr. Art Ary” is planning a small trial to obtain some sound human data on anovel biologic called nissenex, which could reduce percent atheroma volume in patientswith atherosclerosis. Even Dr. Ary is skeptical about nissenex, however, giving it a 2%chance of being truly effective: γ = 0.02. Using a reasonable characterization of the infinitedataset presuming nissenex is really efficacious, the power for the key hypothesis test isjudged to be 0.83 at α = 0.05 and N = 120. Accordingly, the crucial error rates areα∗ = 0.75 and β∗ = 0.004. Thus, three out of four significant tests will be misleading.

Does this high α∗ value imply that the study should not be run? No. If this trial yieldsp ≤ 0.05, it would push this line of research forward to a 1 − 0.75 = 0.25 chance thatnissenex is effective, a major shift from the prior γ = 0.02. If p > 0.05, then there is a1 − 0.004 = 0.996 chance that nissenex has null or near-null efficacy, perhaps solidifying Dr.Ary’s initial skepticism. Thus, either outcome will help Dr. Ary decide whether to continuewith further studies. He also considers using α = 0.20, which gives 0.95 power and makesα∗ = 0.91 and β∗ = 0.001.

1 − α∗ = 0.09 is considerably weaker than the 0.25 computed for α = 0.05, and there islittle practical difference in β∗ values (0.004 versus 0.001). Thus, Dr. Ary will use α = 0.05,but he understands that, given his current prior skepticism regarding the efficacy ofnissenex in treating atherosclerosis, not even a p ≤ 0.05 outcome will sway him toprematurely publicly tout nissenex as effective. It will, of course, encourage him and hissponsors to design and carry out a more confirmatory study. This is prudent scientificpractice. If everyone followed this practice, the scientific literature would not be clutteredwith “significant” findings that fail to replicate in further, larger studies and meta-analyses,provided that any such work takes place (Ioannidis, 2005a, b).

Page 276: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 263

10.9 Table of Crucial Error RatesTable 10.6 shows how α∗ and β∗ depend on γ, α, and β. Type I errors are more frequentearly in the March of Science (low γ), whereas Type II errors are more frequent later in theMarch. Reducing either α or β reduces both α∗ and β∗. Note also that when γ = 0.50 andα = β, then α = α∗ = β = β∗.

Table 10.6 Crucial Type I and Type II Error Rates as a Function of γ, α and Power

α∗: Pr[H0 true | p ≤ α] β∗: Pr[H0 false | p > α]γ : Pr[H0 false] Power β α: 0.01 0.05 0.10 0.20 α: 0.01 0.05 0.10 0.20

0.05 0.30 0.70 .388 .760 .864 .927 .036 .037 .039 .0440.50 0.50 .275 .655 .792 .884 .026 .027 .028 .0320.70 0.30 .213 .576 .731 .844 .016 .016 .017 .0190.80 0.20 .192 .543 .704 .826 .011 .011 .012 .0130.90 0.10 .174 .514 .679 .809 .005 .006 .006 .0070.95 0.05 .167 .500 .667 .800 .003 .003 .003 .003

0.30 0.30 0.70 .072 .280 .438 .609 .233 .240 .250 .2730.50 0.50 .045 .189 .318 .483 .178 .184 .192 .2110.70 0.30 .032 .143 .250 .400 .115 .119 .125 .1380.80 0.20 .028 .127 .226 .368 .080 .083 .087 .0970.90 0.10 .025 .115 .206 .341 .041 .043 .045 .0510.95 0.05 .024 .109 .197 .329 .021 .022 .023 .026

0.50 0.30 0.70 .032 .143 .250 .400 .414 .424 .438 .4670.50 0.50 .020 .091 .167 .286 .336 .345 .357 .3850.70 0.30 .014 .067 .125 .222 .233 .240 .250 .2730.80 0.20 .012 .059 .111 .200 .168 .174 .182 .2000.90 0.10 .011 .053 .100 .182 .092 .095 .100 .1110.95 0.05 .010 .050 .095 .174 .048 .050 .053 .059

0.70 0.30 0.70 .014 .067 .125 .222 .623 .632 .645 .6710.50 0.50 .008 .041 .079 .146 .541 .551 .565 .5930.70 0.30 .006 .030 .058 .109 .414 .424 .438 .4670.80 0.20 .005 .026 .051 .097 .320 .329 .341 .3680.90 0.10 .005 .023 .045 .087 .191 .197 .206 .2260.95 0.05 .004 .022 .043 .083 .105 .109 .115 .127

10.10 SummaryIn writing a single chapter on sample-size analysis, one strives for either breadth or depth.We opted to cover two examples in depth, and thus we failed to even mention any of thevast array of other tools now available to help investigators carefully assess and justifytheir choices for sample sizes across the statistical landscape. What have we not discussed?The list of topics and references is too long to begin and would soon be outdated anyway.

What readers need to understand is that if they have a sample-size analysis issue, theremay be good methodological articles and strategies that address it. If no such help can befound, then Monte Carlo simulation can provide results that are entirely satisfactory. Infact, some excellent statisticians now use simulation for all such problems, even fortraditional ones that have sound “mathematical” solutions that are widely used.

Page 277: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

264 Pharmaceutical Statistics Using SAS: A Practical Guide

We hope the two examples given here provide a rich context to fashion good strategiesto address other problems that may be encountered. Though the methods may vary widely,the core concepts and issues do not.

AcknowledgmentsThe authors thank Drs. Walter Ambrosius, Alex Dmitrienko, and Jean Lightner Norum forskillfully reviewing a draft of this chapter. Comments from other readers are alwayswelcome.

ReferencesAgbenyega, T., Planche, T., Bedu-Addo, G., Ansong, D., Owusu-Ofori, A., Bhattaram, V.A.,

Nagaraja, N.V., Shroads, A.L., Henderson, G.N., Hutson, A.D., Derendorf, H., Krishna, S.,Stacpoole, P.W. (2003). “Population kinetics, efficacy, and safety of Dichloroacetate for lacticacidosis due to severe malaria in children.” Journal of Clinical Pharmacology. 43, 386–396.

Agresti, A. (1980). “Generalized odds ratios for ordinal data.” Biometrics. 36, 59–67.Feynman, R.P. (1999). The Pleasure of Finding Things Out. Cambridge, MA: Perseus Books.Holloway, P.A., Knox, K., Bajaj, N., Chapman, D., White, N.J., O’Brien, R., Stacpoole, P.W.,

Krishna, S. (1995). “Plasmodium Berghei infection: Dichloroacetate improves survival in ratswith lactic acidosis.” Experimental Parasitology. 80, 624–632.

Ioannidis, J.P.A. (2005a). “Contradicted and Initially Stronger Effects in Highly Cited ClinicalResearch.” JAMA. 294, 218–228.

Ioannidis, J.P.A. (2005b). “Why Most Published Research Findings Are False.” PLoS Medicine. 2,696–701.

Johnson, N.L., Kotz, S., Balakrishnan, N. (1994). Continuous Univariate Distributions, Vol. 1 (2ndEd.). New York: John Wiley.

Lee, S.J., Zelen, M. (2000). “Clinical trials and sample size considerations: Another perspective.”Statistical Science. 15, 95–100.

SAS Institute Inc. (2006). SAS/STAT User’s Guide. Cary, NC: SAS Institute Inc.Stacpoole, P.W., Nagaraja, N.V., Hutson, A.D. (2003). “Efficacy of dichloroacetate as a

lactate-lowering drug.” Journal of Clinical Pharmacology. 43, 683–691.Topol, E.J., Califf, R.M., Van de Werf, F., Simoons, M., Hampton, J., Lee, K.L., White, H., Simes,

J., Armstrong, P.W. (1997). “Perspectives on large-scale cardiovascular clinical trials for the newmillennium.” Circulation. 95, 1072–1082.

Wacholder, S., Chanock, S., Garcia-Closas, M., El ghormli, L., Rothman, N. (2004). “Assessing theprobability that a positive report is false: An approach for molecular epidemiological studies.”Journal of the National Cancer Institute. 96, 434–442.

Appendix A Guidelines for “Statistical Considerations” SectionsA well-developed statistical considerations section persuades reviewers that solid skill andeffort have gone into framing the research questions, planning the study, and forming anappropriate team. The writing should be crafted for the clinical researcher who is a good“para-statistician,” as well as for the professional biostatistician. The “StatisticalConsiderations” section should be mostly self-contained and thus may reiterate informationfound elsewhere in the proposal.

A.1 ComponentsDesign. Summarize the study design. It may be helpful to use appropriate terms such asrandomized, double blind, cross-over, controlled, comparative, case-control, prospective,retrospective, longitudinal, and cohort.

Page 278: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 265

Research questions. Strictly speaking, not all studies are driven by testable hypotheses,but all studies have research questions that should be delineated in your Specific Aimssection. Summarize the outcome measures and describe how you expect them to be relatedto the components of the study design and other predictor variables. Restate/translateyour primary research questions into specific estimates of effects and their confidenceintervals, and/or into statistical hypotheses or other methods. Similar descriptionsregarding secondary questions are valuable, too.

Statistical analysis plan. Specify what statistical methods will be used to analyze theprimary and secondary outcome measures. Cite statistical references for non-routinemethods. (Example: The two groups will be compared on KMOB830430 and itsmetabolites using estimates and 95% confidence limits for the generalized odds ratio(Agresti, 1980), which is directly related to the common Wilcoxon rank-sum test.) Thesesections often state what statistical software package and version will be used, but thisusually provides little or no information about what actually will be done.

Randomization (if appropriate). Specify how the randomization will be done,especially if it involves blocking or stratification to control for possible confounding factors.

Sample-size analyses. State the proposed sample size and discuss its feasibility.Estimate the key inferential powers, or other measures of statistical sensitivity/precision,such as the expected widths of key confidence intervals. Strive to make your sample-sizeanalyses congruent with the statistical methods proposed previously, and discuss anyincongruencies. State how you arrived at the conjectures for all the unknowns that underliethe sample-size analysis, citing specific articles and/or summarizing analyses of“preliminary” data or analyses presented in unpublished works. If a sample-size analysiswas not performed, state this categorically and explain why. For example, the proposalmay be only a small pilot study.

Data management. Summarize the schema for collecting, checking, entering, andmanaging the data. What database software will be used? How will the database be tappedto build smaller analysis datasets? Note how you will meet modern standards for datasecurity.

Technical support. Who will perform the necessary database and statistical work? Ifsuch people are less experienced, who will supervise the work?

Appendix B SAS Macro Code to Automate the ProgrammingIn the interest of simplicity, the SAS code provided above avoided all macro programming,except for using the %CrucialRates macro. However, analysts can profit greatly by makingelementary use of the SAS Macro Language. Below is the full program that was used indeveloping the EER example. Note how the parameters that shape the problem arespecified only once at the beginning. Due to rounding, the results obtained with this codediffer slightly from those given above.

options ls=80 nocenter;/************************************************************************\Program Name: EER_SSAnalysis060722.sas

Date: 22 July 2006

Investigator: "Sol Capote, MD; CHI Malaria Research Group"Statistician: "Phynd Gooden, PhD" (Actually, Ralph O’Brien)

Page 279: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

266 Pharmaceutical Statistics Using SAS: A Practical Guide

Purpose: Sample-size analysis for comparing usual care only vs.QCA on elysemine:elysemate ratios at 4 hours (EER4).Assumes data will be logNormal in distribution with samerelative range in the two groups.

\************************************************************************/

*options symbolgen mlogic mprint; * for macro debugging;*options FORMCHAR="|----|+|---+=|-/\<>*"; * for ordinary text tables;

title1 "Usual Care Only (UCO) vs. Usual Care + QCA (QCA)";title2 "Difference in 4-hour elysemine-elysemate ratio (EER4),adjusted";title3 "for three baseline covariates: EER0, plasma lactate, parasitemia";

/*******************************************************************\This program is structured so that all the defining values are setthrough %let macro statements at the start of the code.\*******************************************************************/

/**********************************\BEGIN TECHNICAL SPECIFICATIONS

/**********************************/

* Set label for Y;* ---------------;%let Ylabel = EE Ratio;

* Set variable labels for the two groups ;* ---------------------------------------;%let GrpLabel_1 = UCO;%let GrpLabel_2 = DCA;

/*************************************************************************\Each distribution is logNormal with different medians, but same relativespread (defined below). This is the same as saying that the distributionshave different means but the same coefficients of variation.\*************************************************************************/

* Supply guesstimates for medians ;* ------------------------------- ;%let Ymedian0 = 2.0; * median for control arm, only one scenario ;%let Ymedian1A = 1.8; * median for experimental arm, scenario A ;%let Ymedian1B = 1.7; * median for experimental arm, scenario B ;

* Supply guesstimates for the 95% relative spread, defined below;* ------------------------------------------------------------- ;%let YRelSpread95_1 = 2.5; * YRelSpread95, scenario 1 ;%let YRelSpread95_2 = 3.0; * YRelSpread95, scenario 2 ;

*Set NCovariates and supply guesstimates for PrtlCorr(XXX,logY);* -------------------------------------------------------------;%let NCovariates = 0 3 50; * number of covariates ;%let PrtlCorr_XXXlogY = .2 .35 .50 ; * Multiple partial correlation (R) ;

* between covariates ("XXX") and ;* logY, within treatment groups. ;

* Supply prior probabilities that null is false;

Page 280: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 267

* ---------------------------------------------;%let PriorPNullFalse = .20 .30;

/*******************************\END TECHNICAL SPECIFICATIONS

\*******************************/

/**********************************************************************\=====Notes=====

1. Each distribution is logNormal with different medians, but samerelative spread (defined below). This is the same as saying that thedistributions have different means but the same coefficients ofvariation. Under logNormality, medians are also geometric means.

2. Let Y025, Y500 and Y975 be the 2.5%, 50%, and 97.5% quantiles for Y,i.e., Y500 is the median of Y and Y025 and Y975 are the limits ofthe mid-95% range for Y.

3. Define YRelSpread95 = Y975/Y025 to be the inter-95% relative spread.These relative spreads are taken to be equal in control andexperimental groups.

4. Log(Y025), log(Y500), and log(Y925) are the 2.5% quantile, themedian, and the 97.5% quantiles for logY.

If Y ~ logNormal, then log(Y) ~ Normal, so

mean_logY = median_logY = log(Y500).

Let SD_logY be the standard deviation of logY. Then, log(Y025) andlog(Y925) are each 1.96*SD_log2Y units from mean_logY, so

SD_logY = [log(Y025) - log(Y025)]/(2*1.96),

where 1.96 is the 97.5% quantile (Z975) of the standard Normal,Z ~ N(0,1).

Taking 1.96 to "equal" 2, we have,

SD_logY = [log(Y025) - log(Y025)]/4,

With YRelSpread95 = Y975/Y025, we get,

SD_logY = log(YRelSpread95)/4.

5. It some cases it may be more convenient to use anotherrelative spread, say YRelSpread90 or YRelSpread50. UsingZ900 = 1.65 and Z750 = 0.67, we have

SD_logY = log(YRelSpread90)/(2*1.65)

and

Page 281: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

268 Pharmaceutical Statistics Using SAS: A Practical Guide

SD_logY = log(YRelSpread50)/(2*0.67).

Whereas [log(Y750) - log(Y250)] is the interquartile range for logYYRelSpread50 could be called the interquartile relative range for Y.

6. One can show that the coefficient of variation is

CoefVar_Y = sqrt(exp(SD_logY**2 - 1)).

See page 213 of Johnson, Kotz, Balakrishnan (1994), ContinuousUnivariate Distributions, Vol. I.

7. All logs are taken at base 2, but this choice is irrelevant forsample-size analysis.

\*********************************************************************/

/*********\Main code

\*********/

%let SD_log2Y_1 = %sysevalf(%sysfunc(log2(&YRelSpread95_1))/4);%let SD_log2Y_2 = %sysevalf(%sysfunc(log2(&YRelSpread95_2))/4);

data exemplary;group = "&GrpLabel_1";CellWgt = 1;mean_log2Y_A = log2(&Ymedian0);mean_log2Y_B = log2(&Ymedian0);output;

group = "&GrpLabel_2";CellWgt = 2;mean_log2Y_A = log2(&Ymedian1A);mean_log2Y_B = log2(&Ymedian1B);output;

run;

proc print data=exemplary;run;

proc GLMpower data=exemplary;ODS output output=Ntotals;class group;model mean_log2Y_A mean_log2Y_B = group;weight CellWgt;powerStdDev = &SD_log2Y_1 &SD_log2Y_2NCovariates = &NCovariatesCorrXY = &PrtlCorr_XXXlogYalpha = .01 .05power = 0.95 0.99Ntotal = .;

run;

Page 282: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 269

data Ntotals;set Ntotals;if dependent = "mean_log2Y_A"then comparison = "&Ymedian0 vs &Ymedian1A";

if dependent = "mean_log2Y_B"then comparison = "&Ymedian0 vs &Ymedian1B";

if UnadjStdDev = &SD_log2Y_1then YRelSpread95 = &YRelSpread95_1;

if UnadjStdDev = &SD_log2Y_2then YRelSpread95 = &YRelSpread95_2;

run;

proc tabulate data=Ntotals format=5.0 order=data;format Alpha 4.3 YRelSpread95 3.1;class comparison alpha YRelSpread95 CorrXYNominalPower Ncovariates;

var Ntotal;tableNcovariates="Number of covariates; ",comparison="&Ylabel: "* alpha="Alpha"* NominalPower="Power",

YRelSpread95="95% Relative Spread"* CorrXY="Partial R for Covariates"* Ntotal=""*mean=" "

/rtspace=35;run;

proc GLMpower data=exemplary;ODS output output=powers;class group;model mean_log2Y_A mean_log2Y_B = group;weight CellWgt;powerStdDev = &SD_log2Y_1 &SD_log2Y_2Ncovariates = &NCovariatesCorrXY = &PrtlCorr_XXXlogYalpha = .01 .05Ntotal = 300power = .;

run;

data powers;set powers;if dependent = "mean_log2Y_A"then comparison = "&Ymedian0 vs &Ymedian1A";

if dependent = "mean_log2Y_B"then comparison = "&Ymedian0 vs &Ymedian1B";

if UnadjStdDev = &SD_log2Y_1then YRelSpread95 = &YRelSpread95_1;

if UnadjStdDev = &SD_log2Y_2then YRelSpread95 = &YRelSpread95_2;

if power > .999

Page 283: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

270 Pharmaceutical Statistics Using SAS: A Practical Guide

then power999 = .999;else power999 = power;

run;

proc tabulate data=powers format=4.3 order=data;format Alpha 4.3 YRelSpread95 3.1;class comparison alpha YRelSpread95 CorrXYNtotal Ncovariates;

var power999;tableNtotal="Total Sample Size: "* Ncovariates="Number of covariates; ",

comparison="&Ylabel.: "* alpha="Alpha",YRelSpread95="95% Relative Spread"* CorrXY="Partial R for Covariates"* power999=""*mean=" "

/rtspace=35;run;

%macro CrucialRates (PriorPNullFalse = ,Powers = powers,CrucialErrRates = CrucialErrRates);

/**********************************************************************\Converts Alphas and Powers to Crucial Error Rates-------------------------------------------------

<> PriorPNullFalse= value1 value2 ... value10This is gamma = PriorP[Ho false].

<> Powers= InputDSName"InputDSName" corresponds to ODS output statement in PROC POWERor PROC GLMPOWER, such as

proc power;ODS output output=MoralityPowers;

<> CrucialErrRates= OutputDSName"OutputDSName" is SAS dataset name; default: "CrucialErrRates"

\*********************************************************************/

data &CrucialErrRates;set &Powers;array PrNullFalseV{10} _temporary_ (&PriorPNullFalse);beta = 1 - power;iPNF = 1;do until (PrNullFalseV{iPNF} = .);

gamma = PrNullFalseV{iPNF};/* Compute Crucial Type I error rate */TypeError = "Type I";CrucialRate= alpha*(1 - gamma)/(alpha*(1 - gamma) + (1 - beta)*gamma);

output;/* Compute Crucial Type II error rate */TypeError = "Type II";CrucialRate= beta*gamma/(beta*gamma + (1 - alpha)*(1 - gamma));

output;

Page 284: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 10 Sample-Size Analysis for Traditional Hypothesis Testing: Concepts and Issues 271

iPNF + 1;end;

run;%mend; *CrucialRates;

%CrucialRates ( PriorPNullFalse = &PriorPNullFalse,Powers=powers,CrucialErrRates = CrucRates );

proc tabulate data=CrucRates format=4.3 order=data; *&UniversalText;title3 "Crucial Error Rates for QCA Malaria Trial";format Alpha 4.3 YRelSpread95 3.1;class TypeError gamma comparison alpha YRelSpread95 CorrXYNtotal Ncovariates;

var CrucialRate;tableNtotal="Total Sample Size: "* Ncovariates="Number of covariates; ",

comparison="&Ylabel.: "* YRelSpread95="95% Relative Spread"* CorrXY="Partial R for Covariates",

alpha="Alpha"* gamma="PriorP[Null False]"* TypeError="Crucial Error Rate"* CrucialRate=""*mean=" "

/ rtspace = 32;run;

Page 285: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 286: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11

Design and Analysis ofDose-Ranging Clinical Studies

Alex DmitrienkoKathleen FritschJason HsuStephen Ruberg

11.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 273

11.2 Design Considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 277

11.3 Detection of Dose-Response Trends . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 280

11.4 Regression Modeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

11.5 Dose-Finding Procedures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294

11.6 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 309

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 310

Dose-ranging clinical studies play a very important role in early evaluation of safety andefficacy profiles of experimental drugs. The chapter reviews popular statistical methodsused in dose-response analysis, including trend tests, dose-response modeling, and multipletests for identifying safe and effective doses. The testing and estimation proceduresdiscussed in this chapter are illustrated using examples from dose-ranging clinical trials.

11.1 IntroductionDose-ranging studies are conducted at early stages of drug development (both pre-clinicaland clinical stages) to evaluate safety and efficacy of experimental drugs. A large number ofpublications deal with the analysis of dose-ranging studies designed to test several doses (ordose regimens) of an experimental drug versus a control. This area of research, commonlyreferred to as dose-response analysis, covers the relationship between dose and clinicalresponse.

Alex Dmitrienko is Principal Research Scientist, Global Statistical Sciences, Eli Lilly and Company, USA. Kathleen Fritschis Mathematical Statistician, Division of Biometrics III, Food and Drug Administration, USA. (The views presented in thischapter are those of the author and do not necessarily represent the views of the FDA. Kathleen Fritsch’s contribution tothis chapter was completed prior to her employment with the FDA). Jason Hsu is Professor, Department of Statistics, TheOhio State University, USA. Stephen Ruberg is Director, Global Medical Information Sciences, Eli Lilly and Company,USA.

Page 287: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

274 Pharmaceutical Statistics Using SAS: A Practical Guide

There are several distinct research topics within dose-response analysis (Ruberg, 1995a):

1. Assessment of the dose-related trend in the response variable.2. Estimation of the shape of the dose-response function.3. Determination of optimal doses (including identification of the minimum effective dose,

which is of increasing concern as safety issues arise with marketed products).

The three objectives of dose-response analysis are obviously intertwined; however, it isgenerally prudent to examine them one at a time.

First, before making a decision to invest more research dollars in an experimental drug,drug developers need to ensure that a drug effect, manifested by a positive dose-responserelationship, is present. A positive dose-response relationship, defined as a non-decreasingshape in the response variable from the lowest dose to the highest dose, plays a key role inearly drug evaluation. As pointed out by Kodell and Chen (1991), “evidence of adose-response relationship is taken to be a more compelling finding than evidence of apositive effect that does not appear to be dose-related”.

Once it has been demonstrated that an overall dose-related trend is present, one needsto characterize the dose-response function and determine efficacious and safe doses. Tounderstand the importance of this step, note that a significant overall trend is rarelyaccompanied by significant treatment differences at all dose levels. In most cases,significant treatment differences are observed only at some doses included in a dose-rangingstudy and it is critical to identify the range of doses over which a positive dose-response istruly present. This information is used to determine a therapeutic window (a contiguousinterval within which every dose is safe and effective) and will ultimately guide theselection of doses for registration studies.

Dose-response testing and modeling have received much attention in the clinical trialliterature. For a general overview of issues arising in dose-response studies and a discussionof relevant statistical methods, see Ruberg (1995a, 1995b), Chuang-Stein and Agresti(1997) and Phillips (1997, 1998). Also, Westfall et al. (1999, Section 8.5) provide anoverview of dose-finding methods in clinical trials with a large number of SAS examples.

This chapter summarizes popular statistical methods used in dose-response analysis,e.g., trend tests, dose-response models, and dose-finding strategies based on multiple tests.The statistical procedures introduced in this chapter are illustrated using examples fromdose-ranging clinical trials.

11.1.1 Clinical Trial ExamplesTo illustrate the dose-response tests and models that will be introduced in this chapter, wewill use the following three clinical trials examples. The first one is a cross-over trialrepresentative of trials conducted at early stages of clinical development. The other twotrials use a parallel group design and serve as examples of dose-ranging Phase II or PhaseIII trials.

11.1.2 Clinical Trial in Diabetes Patients (Cross-Over Design)A Phase I study in patients with Type II diabetes was conducted to test four doses of anexperimental drug versus placebo using a cross-over design with two periods. Patientsenrolled into the study were assigned to one of four groups (six patients to each group).Each group of patients received 24-hour placebo and experimental drug infusions on twooccasions. The primary pharmacodynamic objective of the study was to examine effects ofthe selected doses on fasting serum glucose. Specifically, the primary pharmacodynamicendpoint was defined as the fasting serum glucose concentration at the end of a 24-hourinfusion.

Page 288: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 275

Figure 11.1 Fasting serum glucose level in the diabetes trial example

Glu

cose

leve

l (m

g/dL

)

120

140

160

180

Infusion rate (mcg/h)

0 10 20 30 40

Figure 11.1 displays the dose-response relationship observed in the diabetes trial. Themean fasting serum glucose level decreases as the infusion rate increases in a monotonefashion. The mean glucose levels are based on least square means computed from a mixedmodel with a fixed group effect and a random subject effect.

The data set with fasting serum glucose measurements (DIABETES data set) can befound on the book’s companion Web site.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data set used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

11.1.3 Clinical Trial in Patients with Asthma (Parallel Group Design)A Phase II trial in 108 patients with mild to moderate asthma was conducted to comparethree doses of an experimental drug to placebo. The drug was administered daily (50, 250,and 1000 mg/day). The efficacy of the experimental drug was studied using spirometrymeasurements. The primary efficacy endpoint was the forced expiratory volume in onesecond (FEV1).

Figure 11.2 displays the results of the trial. It shows the relationship between the meanFEV1 improvement (estimated using least square means) and daily dose in the asthmatrial. The observed dose-response relationship is not monotone—this is an example of the

Figure 11.2 Mean improvement in FEV1 in the asthma trial example

Cha

nge

in F

EV

1 (L

)

0.0

0.1

0.2

0.3

0.4

Daily dose (mg/day)

0 250 500 750 1000

Page 289: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

276 Pharmaceutical Statistics Using SAS: A Practical Guide

so called umbrella-shaped relationship. The mean FEV1 improvement achieves its maximumat the medium dose (250 mg/day) and then drops to a lower value. The treatment effect atthe high dose is even less than that at the low dose. Note, however, that the non-monotonetrend depicted in Figure 11.2 may be due to sampling variability, and the underlyingdose-response relationship may still be positive. A statistical trend test is required to drawconclusions about the true dose-response curve.

The data set with FEV1 measurements (ASTHMA data set) can be found on the book’scompanion Web site.

11.1.4 Clinical Trial in Patients with Hypertension (Parallel Group Design)Consider a Phase II study in 68 patients with hypertension. The patients were randomlyassigned to receive one of three doses of an antihypertensive drug (10, 20 or 40 mg/day) orplacebo. The primary objective of the study was to examine the effect of the selected doseson diastolic blood pressure (DBP).

Figure 11.3 depicts the relationship between the mean reduction in diastolic bloodpressure (based on least square means) and daily dose of the antihypertensive therapy. Theoverall dose-response trend is positive. The treatment effect seems to achieve a plateau atthe 20 mg/day dose.

Figure 11.3 Mean reduction in diastolic blood pressure in the hypertension trial example

Red

uctio

n in

DB

P (

mm

Hg)

0.0

2.0

4.0

6.0

8.0

Daily dose (mg/day)

0 10 20 30 40

The data set with diastolic blood pressure measurements (HYPERTENSION data set)can be found on the book’s companion Web site.

11.1.5 OverviewAs was stated above, dose-response analysis plays an important role in both pre-clinicaland clinical evaluation of compounds. Although some of the methods discussed below canbe used in toxicological studies, this chapter focuses on a clinical setting which includespharmacological studies and randomized clinical trials. See Chapter 5 for a brief overviewof dose-response approaches used in toxicological studies. Also, this chapter deals mainlywith continuous endpoints (contrast and some other tests can also be used with categoricalvariables). Chuang-Stein and Agresti (1997) give a general discussion of approaches todose-response studies with categorical endpoints.

Section 11.2 outlines general principles that need to be considered in designingdose-ranging trials. The section describes issues arising in the design of dose-ranging studiesand discusses the choice of research hypotheses in trials with negative and positive controls.

Page 290: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 277

Section 11.3 discusses statistical procedures for addressing the first objective ofdose-response analysis, i.e., procedures for testing dose-related trends. The section reviewsboth parametric and non-parametric methods. We also briefly discuss sample sizecalculations in dose-ranging trials based on general contrast tests (see Section 11.3.7).

Section 11.4 deals with the second objective—it briefly introduces regression-basedapproaches to examining the shape of dose-response functions, including methods based onlinear and sigmoid models. Lastly, Section 11.5 considers the third objective ofdose-response analysis and reviews approaches to finding the optimal dose based onmultiple pairwise and treatment-control comparisons. This section introduces multiplicityadjustment procedures that are derived using the principles of closed and partition testing.It is worth noting that, in general, dose-response analyses are performed in Phase II trialsand thus adjustments for multiplicity are not always considered. A strict control of theType I error rate is mandated only in registration trials.

11.2 Design Considerations

11.2.1 Types of Dose-Ranging StudiesTo define dose-ranging studies, we may consider the entire class of studies that involvemultiple doses of an active compound. This broad class includes dose-escalation ordose-titration studies (i.e., the dose could go up or down for the same trial subject),parallel group studies (subjects are randomized to distinct dose groups) or cross-overstudies involving sequences of various dose administrations. When considering the design ofa dose-ranging study, we must first consider patient safety, especially in the early stages ofdrug development. Regardless of how much in vitro or animal testing has been completed,human subjects must be exposed to a new chemical entity on a gradual basis both in termsof the amount of the drug to be administered as well as the duration of exposure.

11.2.2 Dose-Escalation StudiesThe typical dose-ranging trials that are done initially in humans are dose-escalation trialsin which there is a single administration of a very small dose of the drug to a small numberof subjects—usually four to ten volunteers. Doses are escalated, depending on the safetyand pharmacokinetic responses of the patients, in an effort to explore the boundary oftolerability to the drug. Such designs are often replicated using repeated administration ofa fixed dose of the drug for one- or two-week intervals, again to gradually increase exposureand establish tolerable doses of the new compound. It is worth noting that the assignmentof subjects to dose groups in dose-escalation trials is non-random. Subjects are randomizedwithin each dose group but not across the groups as in parallel group designs.

We can consider having the same patients progress through the entire dose escalation orhaving independent groups of volunteers for each step of the way. The choice depends onthe nature of the responses and the objectives of the trial:

• If the response is one that persists, independent groups will be required to avoidprolonged washout periods.

• If the objective is to collect safety information on as many subjects as safely possible,independent groups will be preferred. Of course, using the same patients throughout theentire escalation of doses allows more direct comparisons of responses across doses andthereby more precise estimates of parameters of interest.

• Lastly, in dose-escalation studies, we need to be prepared to deal with incomplete datacaused by dropouts, especially when the dropout is due to intolerance.

Page 291: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

278 Pharmaceutical Statistics Using SAS: A Practical Guide

11.2.3 Cross-Over DesignIn the early stages of drug development, but after tolerability has been established asdescribed above, larger studies with greater sophistication can be initiated. An example ofa cross-over design that embodies elements of randomization and dose escalation is shownin Table 11.1. A very useful aspect of the design is that, in the first period, two-thirds ofthe patients receive the lower dose of the drug. As the periods of the study progress, morepatients are exposed to the higher dose, and all patients are exposed to the lower dosebefore the final period.

Table 11.1 A Cross-Over Study with Three Periods

Sequence Period 1 Period 2 Period 3

1 Placebo Lower dose Higher dose2 Lower dose Placebo Higher dose3 Lower dose Higher dose Placebo

This design is usually appealing to clinicians because of the safety of patients inherentin the dose escalation embedded in the cross-over periods. In addition, the use of placebo ineach period helps maintain the double-blind nature of the trial, which is usually desirable,and allows for the separation of treatment and period effects. All of the usualconsiderations need to be made when considering a cross-over versus parallel design (e.g.,within- and between- subject variability, stability of the disease state).

11.2.4 Parallel Group DesignThe most common and simple design is the controlled, randomized, parallel dose responsestudy. In this study design, patients are randomly allocated to one of several active dosegroups or control, most often placebo. While a placebo control leads to the most clearinterpretation of results, active controls can also be used. The parallel design is mostpopular in Phase II development when larger studies are done in order to explore safetyand effectiveness of a new drug. Since the only difference between treatment groups is thedose of the drug, the design leads to more straightforward analysis and interpretation aswill be described in subsequent sections.

11.2.5 Factorial DesignIn a growing number of situations, researchers want to study the effect of more than oneaspect of the dose of a new compound on a disease state. Several examples come to mind:the amount of the dose and the frequency of dosing (i.e., once a day or twice a day) or thesize of a bolus injection and the subsequent infusion rate of a compound. Furthermore, insome disease states combination therapy is the norm, or at least a common phenomenon,such as chemotherapy and antihypertensive therapy. It may be of interest to study multipledoses of each drug in combination with each other. Obviously, such explorations of doseresponse lead to factorial studies. Statisticians have long recognized the value and economyof conducting factorial experiments, but their use in clinical trials is less common. Thismay be due to the complexity (e.g., packaging and blinding clinical medications) of manyclinical trials. Such designs may be most relevant if one is interested in finding the optimaldosing regimen for a drug or combination of drugs.

11.2.6 Choice of a Control and Research HypothesisNegative and positive controls play a key role in dose-ranging clinical trials. As pointed outin many publications, a significant dose-response trend in the absence of a control groupcannot serve as evidence of a drug effect. In studies with a negative control, a significant

Page 292: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 279

dose-response relationship can be observed even if the lowest dose is less efficacious thanthe control and the highest dose is generally comparable to the control.

Most commonly, dose-ranging clinical trials are designed to investigate the effect ofmultiple doses of an experimental drug compared to a negative control (placebo). Placebois a “dummy” treatment that appears as identical as possible to the test treatment butdoes not contain the test drug. As stated in the (amended) Helsinki Declaration and ICHE10 guidance document, the use of a placebo may be justified in studies where no proventherapeutic method exists. Using a placebo as the control may also be justified if thedisease does not cause serious harm (e.g., seasonal allergy) or when treatment may beoptional (e.g., treatment for pain or depression). However, if the disease causes mortality,the inclusion of a placebo-controlled group may not be ethical. Even if the disease does notcause mortality but causes irreversible morbidity, that is, the disease invariably progressesunless treated (e.g., Type II diabetes), the inclusion of a placebo group may be unethical aswell.

11.2.7 Superiority and Non-Inferiority TestingIn comparing an experimental drug with a negative control, we hope to prove that the newdrug is superior to the negative control by a clinically meaningful amount. If μnegative is themean response of the negative control, and μdrug is the mean of the experimental drug, thenthe drug is considered efficacious if

μdrug > μnegative + δsup,

where δsup is a prespecified non-negative quantity representing a clinically importanttreatment difference for establishing superiority.

An active control is a drug which has been proven to be efficacious, typically alreadyapproved and in use. In comparing a new (experimental) drug against an active control,clinical researchers may hope to prove that the new drug is superior to the active control. Ifμactive is the mean of the active control, then the new drug is considered efficacious if

μnew > μactive + δsup,

where δsup again represents a clinically important treatment difference.This is done primarily in the European Union. The rationale for demanding proven

superiority before approving a drug is that, by limiting the number of drugs and thusallocating each a large share of market, it might be possible to better negotiate acost-effective managed care plan.

Alternatively, clinical researchers may hope to demonstrate that the new drug isnon-inferior to the active control, i.e., show that

μnew > μactive − δnon-inf,

where δnon-inf is the so-called non-inferiority margin that defines a clinically meaningfuldifference in non-inferiority trials.

This approach is often taken in the United States. In such cases, δnon-inf may be afraction (e.g., 50%) of the presumably known improvement the active control provides overthe negative control. For example, in the U.S., a new drug for some mild digestive disordermight be approved if it is shown to maintain at least half the treatment effect of anotherdrug on the market. The rationale for approving such a drug is to let the consumers maketheir own decisions, based on cost, efficacy, and side effect considerations.

In dose-ranging trial with a non-inferiority objective, in addition to the new treatmentand the active control groups, it is desirable to include a negative control group as well.This is done to ensure assay sensitivity. It is best to proceed to compare the new drug withthe active control only after establishing that the non-inferiority trial is sensitive enough to

Page 293: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

280 Pharmaceutical Statistics Using SAS: A Practical Guide

detect the known difference between the active and negative controls. Otherwise, clinicalresearchers may fail to detect a difference between the new treatment and active control(and conclude that the new treatment is non-inferior to the active control) simply due tolack of power. Obviously, if there is a statistically significant difference between the newtreatment and the active control, the difference stands whether the active control is foundto be different from the placebo or not.

11.2.8 One-Sided and Two-Sided TestingOf particular relevance when designing a study to assess a dose-response relationship is thematter of one-sided versus two-sided testing. This issue is debated passionately by some,but it is clear that in the vast majority of situations, the researcher has a known interest inthe direction of the desired response. This would naturally imply a one-sided alternativewith a suitable sample size to carry out the appropriate statistical test. The size of the testalso needs to be considered carefully in the design of the trial.

11.2.9 Control of Type I and Type II Error RatesWhen taking a hypothesis testing approach, we must consider the importance of Type Iand Type II errors. If the response of interest is an efficacy response, then the Type I erroris of greatest interest (i.e., we do not want to conclude a drug is effective when it truly isnot). However, if the response of interest is a safety variable (e.g., QTc prolongation), theType II error plays a more important role (i.e., we do not want to conclude there is noeffect on safety when in fact there is). Often, this is under-appreciated and in the case of asafety study it may be perfectly acceptable to have a larger than usual Type I error ratesuch as 0.10 or 0.15 to shrink the Type II error for a limited or fixed sample size that maybe needed for a practical clinical trial.

11.3 Detection of Dose-Response TrendsIt is commonly agreed that the first step in analyzing dose-response trends and morecomplex problems related to dose-finding is the assessment of the overall effect of anexperimental drug. This step is essentially a screening test to determine whether or not thedrug effect exists at all.

Mathematically, the goal of the overall drug effect assessment is to test the nullhypothesis of no treatment effect. For example, in the diabetes trial example, clinicalresearchers were interested in testing whether or not there was any drug-relatedimprovement in the response variable as the infusion rate increased. The corresponding nullhypothesis of no treatment effect is given by

H0 : μP = μD1 = μD2 = μD3 = μD4,

where μP is the mean glucose level in the placebo period and μD1, μD2, μD3, μD4 are themean glucose levels in the four dosing periods. The null hypothesis of no treatment effect istypically tested against the ordered alternative. Under the ordered alternative, the responseis assumed to increase monotonically with dose:

HA : μP ≤ μD1 ≤ μD2 ≤ μD3 ≤ μD4 and μP < μD4.

In this section we will discuss statistical methods for assessing dose-related trends in theresponse variable. We will begin with simple contrast tests, discuss powerful isotonicmethods (Bartholomew and Williams tests) and introduce a non-parametric approach totesting dose-response trends.

Page 294: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 281

11.3.1 Contrast TestsThe simplest way of testing the null hypothesis of no treatment effect is based on the Ftest associated with a simple ANOVA model. However, the F test is not especially powerfulin a dose-response setting because it does not take advantage of the dose order information.The ANOVA-based approach to examining the overall drug effect can be greatly improvedif we select a contrast that mimics the most likely shape of the dose-response curve andcarries out a trend test associated with this contrast.

To define contrast tests, consider a general dose-ranging study with m doses of a drugtested versus a placebo. Let θ0 be the true value of the response variable in the placebogroup and θ1, . . . , θm denote the true values of the response variable in the m dose groups.The θ parameters can represent mean values of the response variable when it is continuousor incidence rates when the response variable is binary. Further, let θ0, . . . , θm be thesample estimates of the θ parameters in the placebo and dose groups.

A contrast is defined by m + 1 contrast coefficients (one for each treatment group)denoted by

c0m, . . . , cmm.

The coefficients are chosen in such a way that they add up to 0, i.e.,m∑

i=0

cim = 0.

Contrasts are often defined with integer coefficients since the coefficients are standardizedwhen the test statistic is computed.

Once a contrast has been selected, the corresponding t statistic is computed by dividingthe weighted sum of the sample estimates θ0, . . . , θm by its standard error:

t =∑m

i=0 cimθi

SE(∑m

i=0 cimθi

) .

In parallel group studies with an equal number of patients in each group (say, n), thisstatistic follows a t distribution with (m + 1)(n − 1) degrees of freedom.

Popular contrast tests based on linear, modified linear, and maximin contrasts aredefined below.

Linear Contrast

The linear contrast is constructed by assigning integer scores (from 0 to m) to placebo andm ordered doses and then centering them around 0 (Tukey et al., 1985; Rom et al., 1994).In other words, linear contrast coefficients are given by:

cim = i − m/2.

Linear contrast coefficients for dose-ranging studies with 2, 3, and 4 dose groups aredisplayed in Table 11.2.

As was noted above, each contrast roughly mimics the dose-response shape for which itis most powerful. The linear contrast test is sensitive to a variety of positive dose-responseshapes, including a linear dose-response relationship. If the test is significant, this does notnecessarily imply that the true dose-response relationship is linear.

Secondly, the linear contrast introduced above was originally proposed for dose-responsestudies with equally spaced doses and equal sample sizes across the treatment groups.However, the same coefficients are frequently used even when these assumptions areviolated. When the doses are not equally spaced, the linear contrast essentially becomes anordinal contrast that ignores the actual dose levels and replaces them with ordinal scores.

Page 295: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

282 Pharmaceutical Statistics Using SAS: A Practical Guide

Maximin and Modified Linear ContrastsThe maximin contrast was derived by Abelson and Tukey (1963) and possesses aninteresting optimal property. The contrast maximizes the test’s power against theworst-case configuration of the dose effects under the ordered alternative HA:

HA : θ0 ≤ . . . ≤ θm and θ0 < θm.

The maximin contrast is defined as follows:

cim =√

i − i2

m + 1−

√i + 1 − (i + 1)2

m + 1.

Along with the optimal maximin contrast, Abelson and Tukey (1963) also proposed asimple modification of the linear contrast that performs almost as well as the maximincontrast. The modified linear contrast (termed the linear-2-4 contrast by Abelson andTukey) is similar to the linear contrast. The only difference is that the end coefficients arequadrupled and the coefficients next to the end coefficients are doubled. Unlike themaximin coefficients, the modified linear coefficients are easy to compute and remember.

To understand the difference between the linear and maximin tests, note that, unlike thelinear tests, the maximin and modified linear tests assign large weights to the treatmentmeans in the placebo and highest dosing groups. As a result, these tests tend to ignore thedrug effect in the intermediate dosing groups. Although the maximin and modified lineartests are generally more powerful than the simple linear test, they may perform poorly intrials with non-monotonic dose-response curves.

Maximin and modified linear coefficients for dose-ranging studies with 2, 3, and 4 dosegroups are shown in Table 11.2.

Table 11.2 Linear, Modified Linear, and Maximin Contrasts in Dose-RangingStudies with 2, 3, and 4 Dose Groups

Contrast coefficientsContrast Placebo Dose 1 Dose 2 Dose 3 Dose 4

2 dose groups

Linear −1 0 1Modified linear −4 0 4Maximin −0.816 0 0.816

3 dose groups

Linear −3 −1 1 3Modified linear −12 −2 2 12Maximin −0.866 −0.134 0.134 0.866

4 dose groups

Linear −2 −1 0 1 2Modified linear −8 −2 0 2 8Maximin −0.894 −0.201 0 0.201 0.894

Note: The linear and modified linear coefficients have been standardized.

11.3.2 Contrast Tests in the Diabetes Trial ExampleProgram 11.1 analyzes changes in fasting serum glucose levels between the placebo andfour dosing periods in the diabetes trial example. The raw data (DIABETES data set) usedin the program can be found on the book’s companion Web site. To account for the

Page 296: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 283

cross-over design, the program uses the MIXED procedure to fit a simple mixed modelwith a fixed group effect and a random subject effect. The drug effect is assessed using theoverall F test as well as the three contrast tests introduced in this section. The contrastcoefficients included in the three CONTRAST statements are taken from Table 11.2.Lastly, the program uses the Output Delivery System (ODS) to select relevant sections ofthe PROC MIXED output.

Program 11.1 F and contrast tests in the diabetes trial example

proc mixed data=diabetes;ods select tests3 contrasts;class subject rate;model glucose=rate/ddfm=satterth;repeated/type=un subject=subject;contrast "Linear" rate -2 -1 0 1 2;contrast "Modified linear" rate -8 -2 0 2 8;contrast "Maximin" rate -0.894 -0.201 0 0.201 0.894;run;

Output from Program 11.1

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

rate 4 20.2 90.49 <.0001

Contrasts

Num DenLabel DF DF F Value Pr > F

Linear 1 21.3 151.14 <.0001Modified linear 1 21.6 163.63 <.0001Maximin 1 21.6 163.56 <.0001

Output 11.1 lists the test statistics and associated p-values produced by the overall Ftest and the linear, modified linear and maximin tests. The test statistics are very largeand offer strong evidence against the null hypothesis of no drug effect.

11.3.3 Contrast Tests in the Asthma Trial ExampleA similar SAS program can be used to analyze dose-response trends in a parallel groupsetting, e.g., in the asthma trial introduced in Section 11.1. The only change that needs tobe made in Program 11.1 is deleting the REPEATED statement in PROC MIXED.Program 11.2 carries out the overall F test and three contrast tests to analyze meanchanges in FEV1 collected in the asthma trial example. The contrast coefficients for thisfour-arm trial come from Table 11.2. The raw data (ASTHMA data set) used in theprogram can be found on the book’s companion Web site.

Page 297: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

284 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 11.2 F - and contrast tests in the asthma trial example

proc mixed data=asthma;ods select tests3 contrasts;class dose;model change=dose;contrast "Linear" dose -3 -1 1 3;contrast "Modified linear" dose -12 -2 2 12;contrast "Maximin" dose -0.866 -0.134 0.134 0.866;run;

Output from Program 11.2

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

dose 3 104 1.70 0.1707

Contrasts

Num DenLabel DF DF F Value Pr > F

Linear 1 104 1.86 0.1758Modified linear 1 104 1.86 0.1760Maximin 1 104 1.85 0.1764

Output 11.2 shows the test statistics and p-values produced by the overall F test andthe three contrast tests introduced earlier in this section. The four test statistics indicatethat the non-monotone (umbrella-shaped) dose-response relationship depicted inFigure 11.2 is actually consistent with the null hypothesis of no drug effect. Note that, inthe presence of an umbrella-shaped dose-response curve, the contrast test statistics arecomparable in magnitude and are similar to the test statistic of the overall F test.

11.3.4 Isotonic TestsAn alternative approach to testing dose-response trends is based on isotonic methods1.Unlike contrast tests, isotonic tests rely heavily on the assumption of a monotonedose-response relationship. This assumption is reasonable in most dose-ranging studiesbecause higher doses generally produce stronger treatment effects compared to lower doses.

Two most popular isotonic tests were proposed by Bartholomew (1961) and Williams(1971, 1972). Both of them are based on maximum likelihood estimates of the treatmentmeans under the monotonicity constraint and, as a result, are computationally intensive.Due to the computational complexity of the Bartholomew test, this section will focus onthe Williams test for balanced dose-ranging studies with continuous endpoints. It is worthnoting the Williams approach can be extended to test for trends in the binary case(Williams, 1988) or in a non-parametric setting (Shirley, 1977).

The Williams trend test is based on a comparison between the highest dose and placebogroups under the monotonicity constraint. To be precise, the Williams test statistic Wm isa two-sample t statistic in which the sample mean in the highest dose group is replaced

1The word isotonic is used in medical literature to describe muscular contractions. In this context, isotonic refers to amonotonically increasing dose-response function.

Page 298: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 285

with the maximum likelihood estimate under the ordered alternative HA:

HA : μ0 ≤ . . . ≤ μm.

The Wm statistic is given by

Wm =μm − X0

s√

2/n,

where X0 is the regular sample mean of the response variable in the placebo group, μm isthe maximum likelihood estimate of the average response in the highest dose group underthe ordered alternative, s is the pooled sample standard deviation, and n is the sample sizeper group. In the balanced case, the maximum likelihood estimate μm is defined as follows

μm = max[∑m

i=1 Xi

m,

∑mi=2 Xi

m − 1, . . . , Xm

].

Although the Williams statistic is similar to the two-sample t statistic, it no longer followsa t distribution. To find the critical values of the null distribution of Wm, we need to use arather complicated algorithm given in Williams (1971). This algorithm is implemented inthe PROBMC function. Using this function, we have written a macro for carrying out theWilliams test in dose-ranging trials (the %WilliamsTest macro can be found on the book’scompanion Web site).

The %WilliamsTest macro has three arguments described below:

• DATASET is the data set to be analyzed.• GROUP is the name of the group variable in the data set.• VAR is the name of the response variable in the data set.

The macro assumes that the input data set is sorted by the group variable and the firstgroup is the placebo group. Note also that the Williams test was developed for parallelgroup designs and thus the %WilliamsTest macro will not work with cross-over trials.

To illustrate the use of the Williams test, we will apply it to test for a monotonicdose-related relationship in the asthma trial (see Program 11.3).

Program 11.3 Williams test in the asthma trial example

%WilliamsTest(dataset=asthma,group=group,var=change);

Output from Program 11.3

Williams statistic P-value

1.7339 0.0538

Output 11.3 displays the Williams statistic and associated p-value. The p-value ismarginally significant (p = 0.0538) and thus the true dose-response relationship in theasthma trial is unlikely to be positive. Also, it is instructive to compare the p-valueproduced by the Williams test to those produced by popular contrast tests (Output 11.2).The Williams p-value is much smaller than the p-values listed in Output 11.2 whichindicates that in this example the Williams test is more sensitive to the dose-responsetrend than the linear, modified linear, and maximin contrast tests.

Page 299: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

286 Pharmaceutical Statistics Using SAS: A Practical Guide

11.3.5 Jonckheere TestSo far we have discussed dose-response tests for normally or nearly normally distributedendpoints. The assumption of normality may not be met (or may be difficult to justify) insmaller proof-of-concepts trials in which case clinical researchers need to consider anon-parametric test for dose-related trends.

A popular non-parametric trend test for dose-ranging studies was proposed byJonckheere (1954). A similar test was also described by Terpstra (1952) and, for thisreason, the Jonckheere test is sometimes referred to as the Jonckheere-Terpstra test.Another non-parametric trend test was recently proposed by Neuhauser, Liu, and Hothorn(1998). This test is based on a simple modification of Jonckheere’s approach and is morepowerful than the original Jonckheere test. However, the Neuhauser-Liu-Hothorn test is notyet available in SAS.

The Jonckheere test is based on counting the number of times a measurement from onetreatment group is smaller (or larger) than a measurement from another treatment groupand comparing the obtained counts to the counts that would have been observed under thenull hypothesis of no drug effect. To define the Jonckheere approach, let Xij be themeasurement from the jth patient in the ith treatment group, i = 0, . . . , m. The magnitudeof response in two treatment groups, say, kth and lth groups, can be assessed in anon-parametric fashion using the Mann-Whitney statistic Ukl. This statistic is equal to thenumber of times Xkj < Xlj′ plus one-half the number of times Xkj = Xlj′ . The Jonckheerestatistic, defined for all pairwise comparisons as a sum of the Mann-Whitney statistic,combines the information across the m + 1 groups.

T =m∑

k=0

m∑l=k+1

Ukl.

Once the T statistic has been computed, statistical inferences can be performed using anormal approximation or exact methods. First, we can compute a standardized teststatistics which is asymptotically normally distributed and then find a p-value from thisnormal distribution. Alternatively, a p-value can be computed from the exact distributionof the T statistic.

The Jonckheere test is implemented in the FREQ procedure, which supports both theasymptotic and exact versions of this test. The Jonckheere test (in its asymptotic form) isrequested by adding the JT option to the TABLES statement. To illustrate, Program 11.4carries out the Jonckheere test to examine the dose-response relationship in the asthmatrial.

Program 11.4 Jonckheere test in the asthma trial example

proc freq data=asthma;tables dose*change/noprint jt;run;

Page 300: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 287

Output from Program 11.4

Statistics for Table of dose by change

Jonckheere-Terpstra Test

Statistic 2450.5000Z 1.4498One-sided Pr > Z 0.0736Two-sided Pr > |Z| 0.1471

Effective Sample Size = 108Frequency Missing = 4

Output 11.4 shows that the standardized Jonckheere statistic for testing thedose-related trend in FEV1 changes is 1.4498. The associated two-sided p-value is fairlylarge (p = 0.1471), suggesting that the dose-response relationship is not positive.

It is worth noting that PROC FREQ also supports the exact Jonckheere test. Torequest an exact p-value, we need to use the EXACT statement as shown below

proc freq data=asthma;tables dose*change/noprint jt;exact jt;run;

It is important to remember that exact calculations may take a very long time (even inSAS 9.1) and, in some cases, even cause SAS to run out of memory.

11.3.6 Comparison of Trend TestsSeveral authors, including Shirley (1985), Hothorn (1997) and Phillips (1997, 1998),studied the power of popular trend tests in a dose-ranging setting, including contrast tests(linear and maximin), Williams, Bartholomew and Jonckheere tests.

It is commonly agreed that contrast tests can be attractive in dose-ranging trialsbecause they gain power by pooling information across several dose groups. This poweradvantage is most pronounced in cases when the shape of the dose-response curve matchesthe pattern of the contrast coefficients. The contrast tests introduced in this sectionperform best when the response increases with increasing dose. However, when anon-monotonic (e.g., umbrella-shaped) dose-response curve is encountered, contrast testsbecome less sensitive and may fail to detect a dose-response relationship. The same is alsotrue for the Jonckheere test.

The isotonic tests (Williams and Bartholomew tests) tend to be more robust thancontrast tests and perform well in dose-ranging trials with monotonic and non-monotonicdose-response curves (as shown in Output 11.3). To summarize his conclusions about theperformance of various trend tests, Phillips (1998) stated that

specialized tests such as Williams’ and Bartholomew’s tests which are tailored to thespecial ordering of dose groups are powerful against a variety of different patterns ofresults, and therefore, should be used to establish whether or not there is any drugeffect.

11.3.7 Sample Size Calculations Based on Linear Contrast TestsIn this subsection, we will briefly discuss a problem that plays a key role in the design ofdose-ranging trials—calculation of the trial’s size. Consider a clinical trial with a parallel

Page 301: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

288 Pharmaceutical Statistics Using SAS: A Practical Guide

group design in which m doses of an experimental drug are tested against a placebo. Thesame number of patients, n, will be enrolled in each arm of the trial. The total sample size,n(m + 1), is chosen to ensure that, under a prespecified alternative hypothesis, anappropriate trend test will have 1 − β power to detect a positive dose-response relationshipat a significance level α. Here β is the Type II error probability.

Sample size calculations for isotonic tests (Williams and Bartholomew tests) tend to berather complicated, and we will focus on general contrast tests introduced inSection 11.3.1. As before, θ0 will be the true value of the response variable in the placebogroup, and θ1, . . . , θm will denote the true values of the response variable in the m dosegroups (the response can be a continuous variable or proportion). Suppose that clinicalresearchers are interested in rejecting the null hypothesis of no drug effect in favor of

HA : θ0 = θ∗0, θ1 = θ∗

1, . . . , θm = θ∗m,

where θ∗0, θ

∗1, . . . , θ

∗m reflect the assumed magnitude of the response in the placebo and m

dose groups. These values play the role of the so-called smallest clinically meaningfuldifference which is specified when sample size calculations are performed in two-arm clinicaltrials.

The presence of a positive dose-response relationship will be tested using a generalcontrast test with the contrast coefficients c0m, c1m, . . . , cmm. Assume that the standarddeviation of θi is σ/

√n. It is easy to show that, under HA, the test statistic t, defined in

Section 11.3.1, is approximately normally distributed with mean√

n∑m

i=0 cimθ∗i

σ√∑m

i=0 c2im

and standard deviation 1. Therefore, using simple algebra, the sample size in each arm isgiven by

n =σ2(zα/2 + zβ)2 ∑m

i=0 c2im

(∑m

i=0 cimθ∗i )

2 .

EXAMPLE: Clinical trial in patients with hypercholesterolemiaA clinical trial will be conducted to study the efficacy and safety profiles of three doses of acholesterol-lowering drug compared to placebo. The primary trial endpoint is the reductionin LDL cholesterol after a 12-week treatment. Clinical researchers hypothesize that theunderlying dose-response relationship will be linear, i.e.,

HA : θ0 = 0 mg/dL, θ1 = 7 mg/dL, θ2 = 14 mg/dL, θ3 = 21 mg/dL,

where θ0 is the placebo effect and θ1, θ2, and θ3 represent the drug effect in the low,medium, and high dose groups, respectively. The standard deviation of LDL cholesterolchanges is expected to vary between 20 and 25 mg/dL. The dose-ranging trial needs to bepowered at 90% (β = 0.1) with a two-sided significance level of 0.05 (α/2 = 0.025).

Program 11.5 computes the sample size in this dose-ranging trial using the linearcontrast test with c03 = −3, c13 = −1, c23 = 1 and c33 = 3 (see Table 11.2).

Page 302: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 289

Program 11.5 Sample size calculations in the hypercholesterolemia dose-ranging trial

data sample_size;* Mean effects in each arm;theta0=0; theta1=7; theta2=14; theta3=21;* Linear contrast test coefficients;c0=-3; c1=-1; c2=1; c3=3;alpha=0.025; * One-sided significance level;beta=0.1;z_alpha=probit(1-alpha);z_beta=probit(1-beta);do sigma=20 to 25;

sum1=theta0*c0+theta1*c1+theta2*c2+theta3*c3;sum2=c0*c0+c1*c1+c2*c2+c3*c3;n=ceil(sum2*(sigma*(z_alpha+z_beta)/sum1)**2);output;

end;proc print data=sample_size noobs;

var sigma n;run;

Output from Program 11.5

sigma n

20 1821 1922 2123 2324 2525 27

Output 11.5 lists the patient numbers for the selected values of σ. It is important toremember that these numbers define the size of the analysis population. In order tocompute the number of patients that will be enrolled in each treatment group, we need tomake assumptions about the dropout rate. With a 10% drop-out rate, the sample size inthe first scenario (σ = 20) will be 18/0.9 = 20 patients per arm.

11.4 Regression ModelingIn this section we will introduce statistical methods for addressing the second objective ofdose-ranging studies, namely, characterization of the dose-response curve. While in theprevious section we were concerned with hypothesis testing, this section will focus onmodeling dose-response functions and estimation of their parameters. We will considerapproaches based on linear and sigmoid (or Emax) models.

It is important to note that the modeling approaches discussed below can be used tostudy effects of either actual dose or pharmacokinetic exposure parameters (e.g., areaunder the time-plasma concentration curve or maximum plasma concentration) on theresponse variable of interest. When the drug’s distribution follows simple pharmacokineticmodels, e.g., a one-compartmental model with a first-order transfer rate, the exposureparameters are proportional to the dose (this is known as pharmacokineticdose-proportionality). In this case, the two approaches yield similar results. However, in thepresence of complex pharmacokinetics, dose-proportionality may not hold andexposure-response models are more informative than dose-response models.

Page 303: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

290 Pharmaceutical Statistics Using SAS: A Practical Guide

11.4.1 Linear ModelsHere we will consider a simple approach to modeling the dose- or exposure-responserelationship in dose-ranging trials. This approach relies on linear models. Linear modelsoften serve as a reasonable first approximation to an unknown dose-response function andcan be justified in a variety of dose-ranging trials unless the underlying biological processesexhibit ceiling or plateau effects. If a linear model is deemed appropriate, it is easy to fitusing PROC REG (parallel group designs) or PROC MIXED (parallel group or cross-overdesigns).

As an illustration, we will fit a linear model to approximate the relationship between thedrug exposure (measured by the maximum plasma concentration of the study drug, Cmax)and mean fasting glucose level in the diabetes trial. Program 11.6 fits a linearrepeated-measures model to the glucose and Cmax measurements in the Diabetes data setusing PROC MIXED. A repeated-measures model specified by the REPEATED statementis used here to account for the cross-over nature of this trial. The program also computespredicted fasting glucose levels and a confidence band for the true exposure-responsefunction. The predicted values and confidence intervals are requested using theOUTPREDM option in the MODEL statement.

Program 11.6 Linear exposure-response model in the diabetes trial

proc mixed data=diabetes;class subject;model glucose=cmax/outpredm=predict;repeated/type=un subject=subject;

proc sort data=predict;by cmax;

axis1 minor=none value=(color=black)label=(color=black angle=90 ’Glucose level (mg/dL)’)order=(100 to 200 by 20);

axis2 minor=none value=(color=black) label=(color=black ’Cmax (ng/mL)’);symbol1 value=none i=join color=black line=20;symbol2 value=none i=join color=black line=1;symbol3 value=none i=join color=black line=20;symbol4 value=dot i=none color=black;proc gplot data=predict;

plot (lower pred upper glucose)*cmax/overlay frame haxis=axis2 vaxis=axis1;run;quit;

Output from Program 11.6

Solution for Fixed Effects

StandardEffect Estimate Error DF t Value Pr > |t|

Intercept 175.78 2.1279 23 82.61 <.0001cmax -0.03312 0.003048 23 -10.87 <.0001

Output 11.6 lists estimates of the intercept (INTERCEPT) and slope (CMAX) of thelinear exposure-response model produced by PROC MIXED. The intercept represents thebaseline (placebo) effect and the slope indicates the rate at which the mean glucoseconcentration decreases with increasing exposure.

Figure 11.4 depicts the predicted fasting glucose concentration as a function of Cmaxalong with a series of 95% confidence intervals that form a confidence band. It is clear from

Page 304: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 291

Figure 11.4 Fasting glucose levels predicted by a linear model in the diabetes trial

Glu

cose

leve

l (m

g/dL

)

100

120

140

160

180

200

Cmax (pg/mL)

0 500 1000 1500 2000 2500

Figure 11.4 that the linear model provides a fairly poor fit to the fasting glucose data.Most of the dots representing individual glucose measurements lie below the fitted line andoutside the confidence band. The pattern of individual measurements suggests that thetrue exposure-response relationship is likely to be curvilinear.

11.4.2 Sigmoid ModelsWe saw in the previous subsection that linear models may not always provide an adequateapproximation to exposure-response functions. Non-linear models perform better inpractice and are generally more appealing from a clinical perspective. One way to improvethe performance of simple linear models is by applying a non-linear transformation of theindependent variable, e.g., fitting a linear model with a log-transformed AUC or Cmax.Another popular non-linear modeling approach relies on the sigmoid model (also referredto as the Emax model). This model describes the true treatment effect E as a sigmoidfunction of drug exposure d:

E = E0 +Emaxd

γ

dγ + dγ50

.

In this model,

• E0 is the baseline effect at the zero exposure level.• Emax represents the maximum possible effect of the experimental drug on the response

variable relative to baseline.• d50 is the exposure level at half-maximal effect (sometimes called the median effective

dose). It is easy to verify that the treatment effect E is equal to E0 + Emax/2 whend = d50.

• γ determines the steepness of the sigmoid function. The γ parameter, known as the Hillcoefficient in the pharmacodynamic literature, typically ranges between 1 and 3.

In cross-over trials, the introduced sigmoid model needs to be modified to account forrepeated measurements on the same subject. This is achieved by including a random

Page 305: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

292 Pharmaceutical Statistics Using SAS: A Practical Guide

subject term in the model. Program 11.7 uses a sigmoid model with a random subject termto approximate the exposure-response curve in the diabetes trial:

E = E0 − (Emax + η)dd + d50

,

where η is a random subject term and the γ parameter is set to 1 to simplify non-linearmodeling in this small data set. Note that E is a decreasing function of d since theexperimental drug is expected to lower the glucose level. To fit this complex non-linearmodel, the program relies on the NLMIXED procedure, a powerful procedure that supportsa broad class of non-linear models with random effects.

In Program 11.7, the response variable (GLUCOSE) is modeled as a non-linear functionof a fixed exposure effect (CMAX) and a random subject effect (SUB). The subject effectsare assumed to be normally distributed. The SUBJECT option in the RANDOMstatement identifies groups of related observations made on the same individual.

The initial values of the baseline and maximum effects, E0 and Emax, as well as themedian effective dose, d50, were taken from the dose-response function depicted inFigure 11.1. The initial values of both the within-subject (INTRASUBJECT) andbetween-subject (INTERSUBJECT) variances were 1.

Program 11.7 Sigmoid exposure-response model in the diabetes trial

proc nlmixed data=diabetes;ods select ParameterEstimates;parms e0=180 emax=60 d50=900 intrasigma=1 intersigma=1;prediction=e0-(sub+emax)*cmax/(cmax+d50);model glucose~normal(prediction,intersigma);random sub~normal(0,intrasigma) subject=subject;predict e0-emax*cmax/(cmax+d50) out=predict;

proc sort data=predict;by cmax;

axis1 minor=none value=(color=black)label=(color=black angle=90 ’Glucose level (mg/dL)’)order=(100 to 200 by 20);

axis2 minor=none value=(color=black) label=(color=black ’Cmax (ng/mL)’);symbol1 value=none i=join color=black line=20;symbol2 value=none i=join color=black line=1;symbol3 value=none i=join color=black line=20;symbol4 value=dot i=none color=black;proc gplot data=predict;

plot (lower pred upper glucose)*cmax/overlay frame haxis=axis2 vaxis=axis1;

run;quit;

Output from Program 11.7

Parameter Estimates

StandardParameter Estimate Error DF t Value Pr > |t| Alpha

e0 179.36 2.0873 47 85.93 <.0001 0.05emax 87.1425 16.3395 47 5.33 <.0001 0.05d50 899.75 396.02 47 2.27 0.0277 0.05intrasigma 3.0613 148.03 47 0.02 0.9836 0.05intersigma 101.12 24.8511 47 4.07 0.0002 0.05

Page 306: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 293

Parameter Estimates

Parameter Lower Upper Gradient

e0 175.16 183.56 0.000378emax 54.2717 120.01 -0.00006d50 103.05 1696.45 0.001309intrasigma -294.73 300.85 0.003211intersigma 51.1297 151.12 -0.00003

Output 11.6 shows estimates of the parameters of the sigmoid model. The estimated E0,Emax, and d50 parameters are generally close to their initial values. According to the fittedsigmoid model, the experimental drug is theoretically capable of reducing the mean glucoselevel to 92 mg/dL (= 179 − 87). The half-maximal effect is achieved at the median effectivedose (D50) which is equal to 900 mg/dL. The within-subject variance (INTRASUBJECT)is considerably smaller than the between-subject variance (INTERSUBJECT).

Figure 11.5 Fasting glucose levels (mg/dL) predicted by a sigmoid model in the diabetes trial

Glu

cose

leve

l (m

g/dL

)

100

120

140

160

180

200

Cmax (pg/mL)

0 500 1000 1500 2000 2500

Figure 11.5 displays the fasting glucose concentration predicted by the sigmoid modelwith a 95% confidence band. The fitted dose-response curve has a concave shape and fitsthe data well. There is no obvious pattern in the residuals and many observations fallwithin the 95% confidence band around the fitted curve.

The fitted model can be used for an informal determination of doses or drug exposuresto be investigated in Phase II trials (Ruberg, 1995b). Suppose, for example, that theobjective of the diabetes trial was to reduce the glucose concentration to a “normal” leveldefined as 126 mg/dL. We can see from Figure 11.5 that the lower confidence limit is belowthis clinically important threshold when the maximum plasma concentration is greaterthan 1200 pg/mL. Secondly, the mean glucose concentration falls below the 126 mg/dLthreshold around Cmax = 1700 pg/mL. Using this information, we could define theminimally effective exposure level as a value between 1200 and 1700 pg/mL and effectiveexposure levels as values exceeding 1700 pg/mL.

Page 307: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

294 Pharmaceutical Statistics Using SAS: A Practical Guide

11.5 Dose-Finding ProceduresAs was indicated in Section 11.1, once a positive dose-response has been established,clinical researchers are interested in determining the optimal dose range for theirexperimental drug known as the therapeutic window. As an example, the usual adult dosagerange for prescription ibuprofen is 200 to 800 mg three or four times per day, not to exceed3200 mg total daily. In other words, ibuprofen’s therapeutic window extends from 600 mgto 3200 mg per day. The therapeutic window is more conservative for over-the-counter(OTC) versions. The usual adult dosage range for OTC ibuprofen is 200 to 400 mg everyfour to six hours, not to exceed 1200 mg total daily.

The therapeutic window is defined by the minimum effective dose (MED) and maximumtolerated dose (MTD). As the name implies, the MED is chosen to ensure the efficacy ofboth it and all dose levels higher than it (up to the upper limit of the therapeutic window).Likewise, as the maximum dose, an MTD is chosen to ensure the safety of both it and alldose levels lower than it. In the drug approval process, safety (including the determinationof the MTD) is frequently assessed through simple comparisons of adverse events acrosstreatment arms. The Center for Drug Evaluation and Research at the FDA is currentlystudying how to provide a more detailed statistical assessment of the safety profile of adrug by taking into account the time course or other important factors.

In this section, we will concentrate on the statistical determination of the MED indose-ranging studies with a placebo control, given that a safe range of doses has beententatively determined. For more information on simultaneous tests for identifying theMED and MTD, see Bauer, Brannath, and Posch (2001) and Tamhane and Logan (2002).Further, Bauer et al. (1998) describe testing strategies for dose-ranging studies with bothnegative and positive controls.

11.5.1 MED Estimation under the Monotonicity AssumptionThe problem of estimating the MED is often stated as a problem of stepwise multipletesting. Clinical researchers begin with the highest dose or the dose corresponding to thelargest treatment difference and proceed in a stepwise fashion until they encounter anon-significant treatment difference. The immediately preceeding dose is defined as theminimum effective dose (MED).

To simplify the statement of the multiple testing problem arising in MED estimation, itis common to make the following two assumptions known as monotonicity constraints:

• All doses are no worse than placebo.• If Dose k is not efficacious, the lower doses (Doses 1 through k − 1) are not efficacious

either.

These assumptions are reasonable in clinical trials with a positive dose-responserelationship. However, they are not met when the true dose-response function isumbrella-shaped and stepwise tests relying on the monotonicity assumptions break down inthe presence of non-monotone dose-response curves.

Under the monotonicity constraints, the MED estimation problem is easily expressed interms of sequential testing of m null hypotheses associated with the m dose levels. To definethe null hypotheses, assume that the drug’s effect is expressed as a positive shift. Further,the observations, yij , are assumed to be normally distributed and follow an ANOVA model

yij = μi + εij ,

where μ0, . . . , μm are the true treatment means in the placebo and m dose groups and εij ’sare residuals. Given this model, the null hypotheses used in MED estimation are displayed

Page 308: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 295

in Table 11.3. The clinically important difference is denoted by δ. When the clinicallyimportant difference is assumed to be 0, the kth null hypothesis, HM

0k , simplifies to

μ0 = μ1 = . . . = μk.

The first null hypothesis in Table 11.3, HM01 , states that Dose 1 is ineffective, and therefore

its rejection implies that Dose 1 is the MED. Likewise, if HM01 is retained but HM

02 isrejected, Dose 2 is declared the MED, etc.

Table 11.3 Null Hypotheses Used in MED Estimation under theMonotonicity Constraint

Null hypothesis Interpretation

HM01 : μ0 ≤ μ1 < μ0 + δ Dose 1 is ineffective

HM02 : μ0 ≤ μ1, μ2 < μ0 + δ Doses 1 and 2 are ineffective

......

HM0k : μ0 ≤ μ1, . . . , μk < μ0 + δ Doses 1, . . . , k are ineffective

......

HM0m : μ0 ≤ μ1, . . . , μm < μ0 + δ All doses are ineffective

The stepwise testing approach goes back to early work by Tukey, Ciminera, and Heyse(1985), Mukerjee, Robertson, and Wright (1987), Ruberg (1989) and others who proposedmultiple-contrast methods for examining dose-related trends. This section focuses onstepwise contrast tests considered by Tamhane, Hochberg, and Dunnett (1996) andDunnett and Tamhane (1998). We can also construct stepwise testing procedures forestimating the MED using other tests, e.g., isotonic tests described in Section 11.3. Formore information about MED estimation procedures based on the Williams test, seeDunnett and Tamhane (1998) and Westfall et al. (1999, Section 8.5.3).

11.5.2 Multiple-Contrast TestsThe null hypotheses displayed in Table 11.3 will be tested using multiple-contrast tests. It isimportant to understand the difference between multiple-contrast tests introduced here andthe tests discussed in Section 11.3 (known as single-contrast tests). As their name implies,single-contrast tests rely on a single contrast and are used mainly for studying the overalldrug effect. Each of multiple-contrast tests discussed in this subsection actually relies on afamily of contrasts

(c0m(1), . . . , cmm(1)), (c0m(2), . . . , cmm(2)), . . . , (c0m(m), . . . , cmm(m)).

These contrasts are applied in a stepwise manner to test HM01 , . . . , HM

0m. The k nullhypothesis, HM

0k , is tested using the following t statistic

tk =∑m

i=0 cim(k)μi − δ

SE (∑m

i=0 cim(k)μi),

where μ0, . . . , μm are the sample treatment means in the placebo and m dose groups.Assuming n patients in each treatment group, each of the t statistics follows a tdistribution with ν = (m + 1)(n − 1) degrees of freedom.

It is important to note that the contrasts can be constructed on any scale when theclinically significant difference (δ) is 0. The contrast coefficients are automaticallystandardized when the test statistic is computed. However, when δ > 0, the contrasts needto be on the correct scale because the standardization occurs only after δ is subtracted. Toensure the correct scale is used, the contrast coefficients are defined in such a way thatc0m(k) is negative, ckm(k) is positive and the positive coefficients add up to 1.

Popular multiple-contrast tests used in dose-finding are defined below.

Page 309: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

296 Pharmaceutical Statistics Using SAS: A Practical Guide

Pairwise ContrastsThe easiest way to compare multiple dose groups to placebo is to consider all possibledose-placebo comparisons. The associated test is based on a family of pairwise contrasts.The following coefficients are used when the Dose k is compared to placebo:

c0m(k) = −1, ckm(k) = 1

and all other coefficients are equal to 0.

Helmert ContrastsUnlike the pairwise contrast, the Helmert test combines information across several dosegroups. Specifically, when comparing Dose k to placebo, this test assumes that the lowerdoses (Doses 1 through k − 1) are not effective and pools them with placebo. The kthHelmert contrast is defined as follows:

c0m(k) = . . . = c(k−1)m(k) = −1k

, ckm(k) = 1.

The other coefficients are equal to 0. Helmert contrasts are most powerful when the lowerdoses are similar to placebo.

Reverse Helmert ContrastsThe reverse Helmert contrast test is conceptually similar to the regular Helmert test. Thereverse Helmert test combines information across doses by assuming that the lower dosesare as effective as Dose k. Thus, the kth reverse Helmert contrast are given by

c0m(k) = −1, c1m(k) = · · · = ckm(k) =1k

.

The remaining coefficients are equal to 0. Reverse Helmert contrasts are most powerfulwhen the treatment effect quickly plateaus and the larger doses are similar to the highestdose.

Linear Contrasts

The linear contrast test assigns weights to the individual doses that increase in a linearfashion. Dose k is compared to placebo using the following contrast:

c0m(k) = − k

2l, c1m(k) =

1l

(1 − k

2

), . . . , ckm(k) =

k

2l,

where

l =k

4

(k

2+ 1

)if k is even, l =

12

([k

2

]+ 1

)2

if k is odd.

and [k/2] is the largest integer in k/2. As before, the other coefficients are equal to 0.Linear contrasts are most powerful when the MED is near the middle of the range of dosestested in a trial.

Put simply, the Helmert, reverse Helmert and linear contrasts each correspond todifferent shapes of the dose-response curve for which they are most powerful. Figure 11.6displays the treatment effect configurations that lead to the maximum expected value ofthe t statistic for each type of contrast.

Page 310: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 297

Figure 11.6 Treatment effect configurations leading to the maximum expected value of the t statistic underthe monotonicity constraint

� � � �

Helmert contrast

Placebo Dose 1 Dose 2 Dose 3 Dose 4

μ0

μ0 + δ

��

��

��

� � � �

Reverse Helmert contrast

Placebo Dose 1 Dose 2 Dose 3 Dose 4

μ0

μ0 + δ

��

��

��

� �

� � �

Linear contrast

Placebo Dose 1 Dose 2 Dose 3 Dose 4

μ0

μ0 + δ

��

��

��

11.5.3 Closed Testing Procedures Based on Multiple ContrastsIn this section, we will consider the MED estimation problem from a multiple testingperspective and focus on MED estimation procedures that control the overall Type I errorrate. As was pointed out in the introduction, control of the Type I error probability isrequired in registration trials and is less common in Phase II trials.

Two general stepwise procedures for testing HM01 , . . . , HM

0m can be constructed based onthe principle of closed testing proposed by Marcus, Peritz, and Gabriel (1976). Theprinciple has provided a foundation for numerous multiple tests and has found a largenumber of applications in multiplicity problems arising in clinical trials. For example,Kodell and Chen (1991) and Rom, Costello, and Connell (1994) applied the closed testingprinciple to construct multiple tests for dose-ranging studies.

Very briefly, the closed testing principle is based on a hierarchical representation of amultiplicity problem. In general, we need to consider all possible intersections of the nullhypotheses of interest (known as a closed family of hypotheses) and test each intersection atthe same significance level. After that, the results need to be combined to make inferencesabout the original null hypotheses. In this case, the family of null hypotheses defined inTable 11.3 is already a closed family and therefore closed testing procedures have a simplesequentially rejective form.

Page 311: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

298 Pharmaceutical Statistics Using SAS: A Practical Guide

Two closed procedures for estimating the MED are introduced below. The firstprocedure begins with the most significant dose-placebo comparison and then worksdownward. For this reason, this procedure is known as the step-down procedure. Note thatthe idea behind the step-down approach is that the order in which the dose-placebo testsare examined is driven by the data. By contrast, the fixed-sequence procedure requires thatthe testing sequence be specified prior to data analysis.

11.5.4 Step-Down MED Estimation ProcedureConsider the m contrast test statistics, t1, . . . , tm, and order them from the most significantto the least significant. The ordered t statistics will be denoted by

t(1) ≥ . . . ≥ t(m).

The step-down MED estimation procedure is defined below:

• Consider the most significant t statistic, t(1), and compare it to the prespecified criticalvalue (e.g., two-sided 0.05 critical value) derived from the null distribution of t1, . . . , tmwith ν = (m + 1)(n − 1) degrees of freedom and correlation matrix ρ. This critical valueis denoted by c1. If t(1) exceeds c1, the corresponding null hypothesis is rejected and weproceed to the second most significant t statistic.

• The next statistic, t(2), is compared to c2 which is computed from the null distributionof t1, . . . , tm−1 with the same number of degrees of freedom and correlation matrix asabove (c2 is less than c1). If t(2) > c2, reject the corresponding null hypothesis andexamine t(3), etc.

• The step-down procedure terminates as soon as it encounters a null hypothesis whichcannot be rejected.

Let l be the index of the lowest dose which is significantly different from placebo. Whentesting stops, the monotonicity assumption implies that the null hypotheses HM

0l , . . . , HM0m

should be rejected and therefore Dose l is declared the MED.

11.5.5 Fixed-Sequence MED Estimation ProcedureThis procedure relies on the assumption that the order in which the null hypotheses aretested is predetermined. Let t[1], . . . , t[m] denote the a priori ordered t contrast statistics(any ordering can be used as long as it is prespecified). The fixed-sequence procedure isdefined as follows:

• Compare t[1] to the prespecified critical value of the t distribution with ν degrees offreedom (denoted by c). If t[1] is greater than c, the corresponding null hypothesis isrejected and the next test statistic is examined.

• The next test statistic, t[2], is also compared to c and the fixed-sequence procedureproceeds in this manner until it fails to reject a null hypothesis.

The dose corresponding to the last rejected null hypothesis is declared the MED.

The step-down and fixed-sequence procedures protect the Type I error rate with respectto the entire family of null hypotheses shown in Table 11.3, given the monotonicityconstraints. To be more precise, the two procedures control the probability of erroneouslyrejecting any true null hypothesis in the family regardless of which and how many othernull hypotheses are true. This is known as the control of the familywise error rate in thestrong sense.

Page 312: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 299

11.5.6 Pairwise Multiple-Contrast Test in the Hypertension TrialTo illustrate the use of stepwise tests in MED estimation, we will begin with the pairwisecontrast test. Unlike other contrast tests, the pairwise test is easy to implement in practicebecause the associated correlation matrix has a very simple structure. In the balanced case,the correlation between any two pairwise t statistics is 0.5 and, because of this property, wecan carry out the pairwise contrast test by a stepwise application of the well-knownDunnett test.

In the following program, we will apply the step-down and fixed-sequence versions of theDunnett test to find the MED in the hypertension trial. First, Program 11.8 assesses theoverall drug effect in the hypertension trial using the three contrast tests defined in Section11.3.

Program 11.8 Contrast tests in the hypertension trial

proc mixed data=hypertension;ods select contrasts;class dose;model change=dose;contrast "Linear" dose -3 -1 1 3;contrast "Modified linear" dose -12 -2 2 12;contrast "Maximin" dose -0.866 -0.134 0.134 0.866;run;

Output from Program 11.8

Contrasts

Num DenLabel DF DF F Value Pr > F

Linear 1 64 8.61 0.0046Modified linear 1 64 7.70 0.0072Maximin 1 64 7.62 0.0075

Output 11.8 shows the test statistics and p-values of the linear, modified linear, andmaximin tests. All three p-values are very small which indicates that the response improvesin a highly significant manner with increasing dose in the hypertension trial.

Now that a positive dose-response relationship has been established, we are ready for adose-finding exercise. First, we will use the step-down version of the Dunnett test and thenapply the fixed-sequence Dunnett test.

11.5.7 Step-Down Dunnett Test in the Hypertension TrialProgram 11.9 carries out the step-down Dunnett test to determine the MED in thehypertension trial with the clinically important difference δ = 0. The program uses PROCMIXED to compute the t statistics associated with the three dose-placebo comparisons.The statistics are ordered from the most significant to the least significant and compared tosuccessively lower critical values. Specifically, the critical values of the step-down procedureare computed from the Dunnett distribution for the case of three, two, and onedose-placebo comparisons. These critical values are found using the PROBMC function.Note that the PROBMC function assumes a balanced case. Therefore, we computed theaverage number of patients per group in the hypertension trial (17) and plugged thenumber into the well-known formula for calculating degrees of freedom (4(17 − 1) = 64).

Page 313: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

300 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 11.9 Step-down Dunnett test in the hypertension trial

ods listing close;* Compute t statistics for three dose-placebo comparisons;proc mixed data=hypertension;

class dose;model change=dose;lsmeans dose/pdiff adjust=dunnett;ods output diffs=dunnett;

* Order t statistics;proc sort data=dunnett;

by descending tvalue;* Compute critical values (based on Dunnett distribution);data critical;

set dunnett nobs=m;format c 5.2;order=_n_;c=probmc("DUNNETT2",.,0.95,64,m-_n_+1);label c=’Critical value’;

proc print data=critical noobs label;var order dose tvalue c;ods listing;run;

Output from Program 11.9

Criticalorder dose t Value value

1 20 2.80 2.412 40 2.54 2.263 10 1.10 2.00

Output 11.9 shows the t statistics computed in the hypertension trial and the associatedcritical values of the step-down Dunnett test. The dose groups are ordered by their tstatistic and the ORDER variable shows the order in which the doses will be compared toplacebo. The following algorithm is used to determine significance of the t statistics listedin Output 11.9.

• The most significant test statistic (t(1) = 2.80) arises when we compare the medium doseto placebo. This statistic is greater than the critical value (c1 = 2.41) and therefore weproceed to the next dose-placebo comparison.

• The test statistic for the high dose vs. placebo comparison (t(2) = 2.54) also exceeds thecorresponding critical value (c2 = 2.26). Due to this significant result, we will nowcompare the low dose versus placebo.

• Examining the last dose-placebo comparison, we see that the test statistic (t(3) = 1.10)is less than c3 = 2.00.

Since the last dose-placebo comparison did not yield a significant result, the mediumdose (20 mg/day) is declared the MED. The multiple comparisons summarized inOutput 11.9 enable clinical researchers to provide the following characterization of thedose-response function in the hypertension trial. First, as was shown in Output 11.8, theoverall dose-response trend in diastolic blood pressure change is positive and highlysignificant. Further, we have concluded from Output11.9 that the 20 mg/day dose is theMED and thus the positive dose-response trend in the hypertension trial is due tostatistically significant treatment differences at the 20 mg/day and 40 mg/day dose levels.

Page 314: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 301

It is important to remember that the step-down testing approach relies heavily on themonotonicity assumption. When this assumption is not met, the approach may lead toresults that look counterintuitive. Suppose, for example, that we conclude significance forthe low and medium doses but not for the high dose. In this case, the low dose is declaredthe MED even though we did not actually reject the null hypothesis of no drug effect atthe high dose.

11.5.8 Fixed-Sequence Pairwise Test in the Hypertension TrialProgram 11.10 takes a different approach to the dose-finding problem in the hypertensiontrial. It identifies the MED using the fixed-sequence version of the Dunnett test. Tounderstand the difference between the two approaches, recall that the step-down procedure(see Output 11.9) compares ordered t statistics to successively lower Dunnett critical valueswhereas the fixed-sequence procedure compares the same t statistics to a constant criticalvalue derived from a t distribution (it is the two-sided 95th percentile of the t distributionwith 64 degrees of freedom). Additionally, the fixed-sequence procedure requires that thethree tests for individual dose-placebo comparisons be ordered before the dose-findingexercise begins. We will assume a natural testing sequence here: high dose vs. placebo,medium dose vs. placebo, and low dose vs. placebo.

Program 11.10 Fixed-sequence pairwise test in the hypertension trial

ods listing close;* Compute t statistics for three dose-placebo comparisons;proc mixed data=hypertension;

class dose;model change=dose;lsmeans dose/pdiff adjust=dunnett;ods output diffs=dunnett;

* Prespecified testing sequence;data dunnett;

set dunnett;if dose=10 then order=3;if dose=20 then order=2;if dose=40 then order=1;

* Order t statistics;proc sort data=dunnett;

by order;* Compute critical values (based on t distribution);data critical;

set dunnett;format c 5.2;c=tinv(0.975,64);label c=’Critical value’;

proc print data=critical noobs label;var order dose tvalue c;ods listing;run;

Output from Program 11.10

Criticalorder dose t Value value

1 40 2.54 2.002 20 2.80 2.003 10 1.10 2.00

Page 315: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

302 Pharmaceutical Statistics Using SAS: A Practical Guide

Output 11.10 lists t statistics for the three dose-placebo comparisons and criticalvalues of the fixed-sequence procedure. Note that the testing sequence is no longerdata-driven—the doses are ordered according to the prespecified rule (the ORDER variableindicates the order in which the doses will be compared to placebo). Also, the criticalvalues in Output 11.10 are uniformly smaller than those shown in Output 11.9 with theexception of the low dose vs. placebo test. This means that the fixed-sequence dose-findingprocedure is likely to find more significant differences than the step-down procedure,provided that the prespecified testing sequence is consistent with the true dose-responsecurve.

Proceeding in a stepwise fashion, we see that the test statistics associated with the twohighest doses (2.54 and 2.80) are greater than the critical value. However, the low dose isnot significantly different from placebo because its t statistic is too small. Given thisconfiguration of t values, we conclude that the medium dose (20 mg/day dose) is the MED.When we compare this to Output 11.9, it is easy to see that the two MED estimationprocedures resulted in the same minimally effective dose. However, the conclusions are notguaranteed to be the same, especially when the dose-response curve is not perfectlymonotone.

11.5.9 Other Multiple-Contrast Tests in the Hypertension TrialThe dose-finding procedures described so far relied on pairwise comparisons. Intuitively,these procedures are likely to be inferior (in terms of power) to procedures that poolinformation across dose levels. For example, consider procedures based on a stepwiseapplication of the Helmert or linear contrasts. In general, the problem of computing criticalvalues for the joint distribution of t statistics in stepwise procedures presents a seriouscomputational challenge, especially in the case of unequal sample sizes. This is due to thecomplex structure of associated correlation matrices. Although exact numerical methodshave been discussed in the literature (see, for example, Genz and Bretz, 1999),simulation-based approaches are generally more flexible and attractive in practice. Here wewill consider a simulation-based solution proposed by Westfall (1997).

Westfall (1997) proposed a Monte Carlo method that can be used in dose-findingprocedures based on non-pairwise contrasts. Instead of computing critical values for tstatistics as was done in Program 11.9, the Monte Carlo algorithm generates adjustedp-values. The adjusted p-values are then compared to a prespecified significance level (e.g.,two-sided 0.05 level) to test the null hypotheses listed in Table 11.3. The algorithm isimplemented in the %SimTests macro described in Westfall et al. (1999, Section 8.6).2

Program 11.11 calls the %SimTests macro to carry out the reverse Helmertmultiple-contrast test in the hypertension trial. Reverse Helmert contrasts were chosenbecause they perform well in clinical trials in which the treatment effect reaches aplateau and other multiple-contrast tests can be used if a different dose-relationship isexpected.

As shown in the program, to invoke the %SimTests macro, we need to define severalparameters, including the family of reverse Helmert contrasts, the treatment means in thehypertension trial, and their covariance matrix. The parameters are specified in the%Estimates and %Contrasts macros using SAS/IML syntax. Once these parameters havebeen defined, the macro approximates adjusted two-sided p-values for the threedose-placebo tests using 100,000 simulations.

2As this book was going to press, the methods in the %SimTests and %SimIntervals macros were being completely hard-coded in the GLIMMIX procedure in SAS/STAT software. The methodology is described in Westfall and Tobias (2006).

Page 316: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 303

Program 11.11 Reverse Helmert multiple-contrast test in the hypertension trial

ods listing close;* Compute treatment means and covariance matrix;proc mixed data=hypertension;

class dose;model change=dose;lsmeans dose/cov;ods output lsmeans=lsmeans;run;

%macro Estimates;use lsmeans;* Treatment means;read all var {estimate} into estpar;* Covariance matrix;read all var {cov1 cov2 cov3 cov4} into cov;* Degrees of freedom;read point 1 var {df} into df;

%mend;%macro Contrasts;

* Reverse Helmert contrasts;c={1 -1 0 0, 1 -0.5 -0.5 0, 1 -0.3333 -0.3333 -0.3334};c=c‘;* Labels;clab={"Dose 1 vs Placebo", "Dose 2 vs Placebo", "Dose 3 vs Placebo"};

%mend;* Compute two-sided p-values using 100,000 simulations;%SimTests(nsamp=100000,seed=4533,type=LOGICAL,side=B);proc print data=SimTestOut noobs label;

var contrast adjp seadjp;format adjp seadjp 6.4;ods listing;run;

Output from Program 11.11

SEAdjContrast AdjP P

Dose 1 vs Placebo 0.2753 0.0000Dose 2 vs Placebo 0.0425 0.0002Dose 3 vs Placebo 0.0200 0.0002

Output 11.11 lists Monte Carlo approximations to the adjusted p-values for the threedose-placebo comparisons as well as associated standard errors. The standard errors arequite small which indicates that the approximations are reliable. Comparing the computedp-values to the 0.05 threshold, we see that the highest two doses are significantly differentfrom placebo, whereas the low dose not separate from placebo. As a consequence, themedium dose (20 mg/day dose) is declared the MED. This conclusion is consistent with theconclusions we reached when applying the pairwise multiple-contrast test.

11.5.10 Limitations of Tests Based on the Monotonicity AssumptionIt is important to remember that the dose-finding procedures described in the previoussubsection control the Type I error rate only if the true dose-response relationship ismonotone. When the assumption of a monotonically increasing dose-response relationship

Page 317: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

304 Pharmaceutical Statistics Using SAS: A Practical Guide

is not met, the use of multiple-contrast tests can result in a severely inflated probability offalse-positive outcomes (Bauer, 1997; Bretz, Hothorn and Hsu, 2003). This phenomenonwill be illustrated below.

Non-monotone dose-response shapes, known as the U-shaped or umbrella-shaped curves,are characterized by a lower response at higher doses (see, for example, Figure 11.1). Whenthe dose-response function is umbrella-shaped and the true treatment difference at thehighest dose is not clinically meaningful (i.e., μm − μ0 < δ), the highest dose should not bedeclared effective. However, if the remaining portion of the dose-response function ismonotone, some reasonable tests that control the Type I error rate under the monotonicityconstraint may finally declare the highest dose effective with a very high probability.

11.5.11 Hypothetical Dose-Ranging Trial ExampleTo illustrate, consider a hypothetical trial in which five doses of an experimental drug arecompared to placebo. Table 11.4 shows the true treatment means μ0, μ1, μ2, μ3, μ4, and μ5as well as true standard deviation σ (the standard deviation is chosen in such a way thatσ/

√n = 1). Assume that the clinically important treatment difference δ is 0.

Table 11.4 Hypothetical Dose-Ranging Trial

Treatment groupPlacebo Group 1 Group 2 Group 3 Group 4 Group 5

n 5 5 5 5 5 5Mean 0 0 0 2.5 5 0SD 2.236 2.236 2.236 2.236 2.236 2.236

It can be shown that the sequential test based on pairwise contrasts (i.e., stepwiseDunnett test) controls the familywise error rate under any configuration of true treatmentmeans. However, when other contrast tests are carried out, the familywise error ratebecomes dependent on the true dose-response shape and can become considerably inflatedwhen an umbrella-shaped dose-response function (similar to the one shown in Table 11.4)is encountered.

Consider, for example, the linear contrast test. If linear contrasts are used for detectingthe MED in this hypothetical trial, the probability of declaring Dose 5 efficacious will be0.651 (using a t test with 24 error degrees of freedom). To see why the linear contrast testdoes not control the Type I error rate, recall that, in testing HM

05 defined in Table 11.3,positive weights are assigned to the treatment differences μ4 − μ1 and μ3 − μ2. Theexpected value of the numerator of the t-statistic for testing H05 is given by(

59(0) +

39(5) +

19(2.5)

)−

(19(0) +

39(0) +

59(0)

)= 1.94.

As a result, the test statistic has a non-central t distribution with a positive non-centralityparameter and thus the linear contrast test will reject the null hypothesis HM

05 more oftenthan it should.

The reverse Helmert contrast test, like the linear contrast test, can also have too high anerror rate when the true dose-response curve is umbrella-shaped. In the hypothetical trialexample, the probability of declaring the highest dose efficacious will be 0.377 (again, basedon a t test with 24 degrees of freedom). This error rate is clearly much higher than 0.05.Combining the signal from the third and fourth doses into the estimate μ5 causes thereverse Helmert contrast test to reject the hypothesis too often.

The regular Helmert contrast test does control the Type I error rate in studies withumbrella-shaped dose-response curves. However, it can have an inflated Type I error rateunder other configurations. In particular, the Type I error probability can be inflated if the

Page 318: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 305

response at lower doses is worse than the response observed in the placebo group.Figure 11.7 shows the general shapes of treatment effect configurations that can lead toinflated Type I error rates for each type of contrast.

Figure 11.7 Treatment effect configurations that can lead to inflated Type I error rates

� �

Helmert contrast

Placebo Dose 1 Dose 2 Dose 3 Dose 4

μ0

μ0 + δ

������ ������������

Reverse Helmert contrast

Placebo Dose 1 Dose 2 Dose 3 Dose 4

μ0

μ0 + δ

��

��

��������������

��

��

��

Linear contrast

Placebo Dose 1 Dose 2 Dose 3 Dose 4

μ0

μ0 + δ

��������������

��

����

��

��

The choice of which contrast test to use in dose-finding problems is thus clear:

• If a monotonically increasing dose-response relationship can be assumed, clinicalresearchers need to choose contrasts according to which dose-response shape inFigure 11.6 is expected.

• If a monotone dose-response relationship cannot be assumed, we must choose pairwisecontrasts to protect the overall Type I error probability.

11.5.12 MED Estimation in the General CaseIn this section we will discuss a dose-finding approach that does not rely on the assumptionof a monotone dose-response relationship.

The closed testing principle is widely used in a dose-ranging setting to generate stepwisetests for determining optimal doses. Here we will focus on applications of another powerfulprinciple known as the partitioning principle to dose finding. The advantage of thepartitioning principle is two-fold. First, as shown by Finner and Strassburger (2002), thepartitioning principle can generate multiple tests at least as powerful as closed tests.

Page 319: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

306 Pharmaceutical Statistics Using SAS: A Practical Guide

Secondly, the partitioning principle makes drawing statistical inferences relatively clear-cut:the statistical inference given is simply what is consistent with the null hypotheses whichhave not been rejected (Stefansson, Kim, and Hsu, 1988; Hsu and Berger, 1999).

The partitioning principle is based on partitioning the entire parameter space intodisjoint null hypotheses which correspond to useful scientific hypotheses to be proved.Since these null hypotheses are disjoint, exactly one of them is true. Therefore, testing eachnull hypothesis at a prespecified α level, e.g., α = 0.05, controls the familywise error rate inthe strong sense without any multiplicity adjustments.

To demonstrate how the partitioning principle can be used in dose finding, consider thedose-ranging study introduced earlier in this section. This study is conducted to test mdoses of a drug versus placebo. Assume that positive changes in the response variablecorrespond to a beneficial effect. Table 11.5 shows a set of partitioning hypotheses that canbe used in finding the MED.

Table 11.5 Partitioning Hypotheses for Finding the MED

Null hypothesis Interpretation

HP00 : μ0 + δ < μ1, . . . , μm All doses are effective

HP01 : μ1 ≤ μ0 + δ < μ2, . . . , μm Doses 2, . . . , m are effective but Dose 1 is ineffective

......

HP0k : μk ≤ μ0 + δ < μk+1, . . . , μm Doses k + 1, . . . , m are effective but Dose k is ineffective

......

HP0m : μm ≤ μ0 + δ Dose m is ineffective

It is easy to check that the null hypotheses displayed in Table 11.5 are mutually exclusiveand only one of them can be true. For example, if HP

0m is true (Dose m is ineffective), HP0k

(Doses k + 1, . . . , m are effective but Dose k is ineffective) must be false. Because of thisinteresting property, testing each null hypothesis at a prespecified α level controls thefamilywise error rate in the strong sense at the α level. Note that the last hypothesis, HP

00,does not really need to be tested to determine which doses are effective; however, it isuseful toward pivoting the tests to obtain a confidence set. Secondly, the union of thehypotheses is the entire parameter space and thus inferences resulting from testing thesehypotheses are valid without any prior assumption on the shape of the dose-response curve.

As the null hypotheses listed in Table 11.5 partition the entire parameter space, drawinguseful inferences from testing HP

01, . . . , HP0m is relatively straightforward. We merely state

the inference which is logically consistent with the null hypotheses that have not beenrejected. Consider, for example, the hypertension trial in which four doses were testedagainst placebo (m = 4). The union of HP

04 (Dose 4 is ineffective) and HP03 (Dose 4 is

effective but Dose 3 is ineffective) is “either Dose 3 or Dose 4 is ineffective”. Thus, therejection of HP

03 and HP04 implies that both Dose 3 and Dose 4 are effective.

In general, if HP0k, . . . , H

P0m are rejected, we conclude that Doses k, . . . , m are all

efficacious because the remaining null hypotheses in Table 11.5, namely, HP01, . . . , H

P0(k−1)

are consistent with this inference. As a consequence, Dose k is declared the MED.

11.5.13 Shortcut Testing ProcedureThe arguments presented in the previous section lead to a useful shortcut procedure fordetermining the MED in dose-ranging trials. The shortcut procedure is defined in Table11.6.

The dose-finding procedure presented in Table 11.6 is equivalent to the fixed-sequenceprocedure based on pairwise comparisons of individual doses to placebo (see Section11.5.6). The SAS code for implementing this procedure can be found in Program 11.10.

Page 320: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 307

Table 11.6 Shortcut Testing Procedure for Finding the MED (All null hypotheses are tested at a prespecifiedα level such as a two-sided 0.05 level)

Step Null hypothesis Decision rule

1 HS0m : μm ≤ μ0 + δ If retained, there are no effective doses.

If rejected, infer μm > μ0 + δ and go to Step 2.2 HS

0(m−1) : μm−1 ≤ μ0 + δ If retained, Dose m is the MED.If rejected, infer μm−1 > μ0 + δ and go to Step 3.

......

...k HS

0k : μk ≤ μ0 + δ If retained, Dose k + 1 is the MED.If rejected, infer μk > μ0 + δ and go to Step k + 1.

......

...m HS

01 : μ1 ≤ μ0 + δ If retained, Dose 2 is the MED.If rejected, infer μ1 > μ0 + δ and Dose 1 is the MED.

It is important to remember that the parameter space should be partitioned in such away that dose levels expected to show a greater response are tested first and the set of doselevels inferred to be efficacious is contiguous. For example, suppose that the dose-responsefunction in a study with four active doses is expected to be umbrella-shaped. In this casewe might partition the parameter space so that dose levels are tested in the followingsequence: Dose 3, Dose 4, Dose 2, and Dose 1. However, it is critical to correctly specify thetreatment sequence. It is shown in Bretz, Hothorn, and Hsu (2003) that the shortcutprocedure that begins at the highest dose suffers a significant loss of power when thedose-response function is not monotone.

11.5.14 Simultaneous Confidence IntervalsAnother important advantage of using the partitioning approach is that, unlike closedtesting procedures, partitioning procedures are easily inverted to derive simultaneousconfidence intervals for the true treatment means or proportions. For example, Hsu andBerger (1999) demonstrated how partitioning-based simultaneous confidence sets can beconstructed for multiple comparisons of dose groups to a common control.

To define the Hsu-Berger procedure for computing stepwise confidence intervals,consider the ANOVA model introduced in the beginning of Section 11.5 and let

lk = yk − y0 − tα,νs√

2/n

be the lower limit of the naive one-sided confidence interval for μk − μ0. Here s is thepooled sample standard deviation and tα,ν is the upper 100αth percentile of the tdistribution with ν = 2(n − 1) degrees of freedom, and n is the number of patients pergroup. The naive limits are not adequate in the problem of multiple dose-placebocomparisons because their simultaneous coverage probability is less than its nominal value,100(1 − α)%. To achieve the nominal coverage probability, the confidence limits l1, . . . , lmneed to be adjusted downward. The adjusted lower limit for the true treatment differenceμk − μ0 will be denoted by l∗k.

Assume that the doses will be compared with placebo beginning with Dose m, i.e., Dosem vs. placebo, Dose m − 1 vs. placebo, etc. As before, any other testing sequence can beused as long as it is not data-driven. A family of one-sided stepwise confidence intervalswith a 100(1 − α)% coverage probability is defined in Table 11.7.

Program 11.12 uses the Hsu-Berger method to compute one-sided confidence intervalswith a simultaneous 95% coverage probability for the treatment means in the hypertension

Page 321: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

308 Pharmaceutical Statistics Using SAS: A Practical Guide

Table 11.7 Simultaneous Confidence Intervals in Dose-Ranging Trials

Step Adjusted confidence limits

1 If lm > δ, let l∗m = δ and go to Step 2. Otherwise, let l∗m = lm and stop.2 If lm−1 > δ, let l∗m−1 = δ and go to Step 3.

Otherwise, let l∗m−1 = lm−1 and stop....

...k If lm−k+1 > δ, let l∗m−k+1 = δ and go to Step k + 1.

Otherwise, let l∗m−k+1 = lm−k+1 and stop....

...m + 1 If all doses are efficacious, let l∗k = min(l1, . . . , lm), k = 1, . . . , m.

Note: If Dose k is not efficacious, the adjusted confidence limits l∗1 , . . . , l∗k−1 are not defined.

study3. As in Program 11.10, we will consider a testing sequence that begins at the highdose (high dose vs. placebo, medium dose vs. placebo, and low dose vs. placebo). Also, theclinically important reduction in diastolic blood pressure will be set to 0 (δ = 0).

Program 11.12 Simultaneous confidence intervals in the hypertension trial

ods listing close;* Compute mean squared error;proc glm data=hypertension;

class dose;model change=dose;ods output FitStatistics=mse(keep=rootmse);

* Compute n and treatment mean in placebo group;proc means data=hypertension;

where dose=0;var change;output out=placebo(keep=n1 mean1) n=n1 mean=mean1;

* Compute n and treatment mean in dose groups;proc means data=hypertension;

where dose>0;class dose;var change;output out=dose(where=(dose^=.)) n=n2 mean=mean2;

* Prespecified testing sequence;data dose;

set dose;if _n_=1 then set mse;if _n_=1 then set placebo;if dose=10 then order=3;if dose=20 then order=2;if dose=40 then order=1;

* Order doses;proc sort data=dose;

by order;

3Program 11.12 is based on SAS code published in Dmitrienko et al. (2005, Section 2.4).

Page 322: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 309

data dose;set dose;retain reject 1;format lower adjlower 5.2;delta=0;lower=mean2-mean1-tinv(0.95,n1+n2-2)*sqrt(1/n1+1/n2)*rootmse;if reject=0 then adjlower=.;if reject=1 and lower>delta then adjlower=delta;if reject=1 and lower<=delta then do; adjlower=lower; reject=0; end;label lower=’Naive 95% lower limit’

adjlower=’Adjusted 95% lower limit’;proc print data=dose noobs label;

var order dose lower adjlower;ods listing;run;

Output from Program 11.12

Naive 95% Adjustedlower 95% lower

order dose limit limit

1 40 1.82 0.002 20 2.33 0.003 10 -1.26 -1.26

Output 11.12 displays the naive and adjusted confidence limits for the mean reductionin diastolic blood pressure in the three dose groups compared to placebo as well as theorder in which the doses are compared to placebo (ORDER variable). As was explainedabove, adjusted limits that result in a confidence region with a 95% coverage probabilityare computed in a stepwise manner. The lower limits of the naive one-sided confidenceinterval for the high and medium doses are greater than the clinically important differenceδ = 0 and thus the corresponding adjusted limits are set to 0. The mean treatmentdifference between the low dose and placebo is not significant (the naive confidence intervalcontains 0) and therefore the adjusted limit is equal to the naive limit. If the hypertensiontrial included more inefficient dose groups, the adjusted confidence limits for those doseswould remain undefined.

11.6 SummaryThis chapter reviews most important issues arising in the analysis of dose-ranging trials.The following topics are discussed in the chapter.

• Assessment of the dose-related trend. Dose-response analysis begins with an overallassessment of dose-related trends in the response variable. The chapter reviewscontrast-based, isotonic (Bartholomew and Williams test), and non-parametric(Jonckheere test) approaches to testing dose-response trends.

• Estimation of the shape of the dose-response function. The next step after theassessment of the overall drug effect is the characterization of the underlyingdose-response relationship. This is achieved by modeling dose-response functions andestimation of their parameters The chapter introduces two dose-response models (linearand sigmoid) that are widely used in dose-ranging trials.

• Determination of the optimal dose. The last step in dose-response analysis is thedetermination of the therapeutic window defined by the minimum effective and maximum

Page 323: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

310 Pharmaceutical Statistics Using SAS: A Practical Guide

tolerated doses. Two popular approaches to the estimation of the minimum effectivedose (MED) are considered in the chapter. The first one, based on the principle of closedtesting, relies on a sequential application of contrast tests to determine the smallest dosethat is significantly different from placebo. The closed testing procedures control theoverall Type I error rate only if the underlying dose-response function is monotone. Analternative approach which relies on the partitioning principle can be safely used evenwhen the assumption of monotonicity is not met.

Statistical methods introduced in this chapter are illustrated using examples fromcross-over and parallel group clinical trials.

ReferencesAbelson, R.B., Tukey, J.W. (1963). “Efficient utilization of non-numerical information in

quantitative analysis: General theory and the case of simple order.” The Annals of MathematicalStatistics. 34, 1347–1369.

Bartholomew, D.J. (1961). “Ordered tests in the analysis of variance.” Biometrika. 48, 325–332.Bauer, P. (1997). “A note on multiple testing procedures in dose finding.” Biometrics. 53,

1125–1128.Bauer, P., Brannath, W., Posch, M. (2001). “Multiple testing for identifying effective and safe

treatments.” Biometrical Journal. 43, 605–616.Bauer, P., Rohmel, J., Maurer, W., Hothorn, L. (1998). “Testing strategies in multi-dose

experiments including active control.” Statistics in Medicine. 17, 2133–2146.Bretz, F., Hothorn, L.A., Hsu, J.C. (2003). “Identifying effective and/or safe doses by stepwise

confidence intervals for ratios.” Statistics in Medicine. 22, 847–858.Chuang-Stein, C., Agresti, A. (1997). “A review of tests for detecting a monotone dose-response

relationship with ordinal response data.” Statistics in Medicine. 16, 2599–2618.Dmitrienko, A., Molenberghs, G., Chuang-Stein, C., Offen, W. (2005). Analysis of Clinical Trials

Using SAS: A Practical Guide. Cary, NC: SAS Institute Inc.Dunnett, C.W., Tamhane, A.C. (1998). “Some new multiple-test procedures for dose finding.”

Journal of Biopharmaceutical Statistics. 8, 353–366.Finner, H., Strassburger, K. (2002). “The partitioning principle: a powerful tool in multiple decision

theory.” Annals of Statistics. 30, 1194–1213.Genz, A., Bretz, F. (1999). “Numerical computation of the multivariate t probabilities with

application to power calculation of multiple contrasts.” Journal of Statistical Computation andSimulation. 63, 361–378.

Hothorn, L.A. (1997). “Modifications of the closure principle for analyzing toxicological studies.”Drug Information Journal. 30, 403–412.

Hsu, J.C., Berger, R.L. (1999). “Stepwise confidence intervals without multiplicity adjustment fordose-response and toxicity studies.” Journal of the American Statistical Association. 94, 468–482.

Jonckheere, A.R. (1954). “A distribution-free K sample test against ordered alternatives.”Biometrika. 41, 133–145.

Kodell, R.L., Chen, J.J. (1991). “Characterization of dose-response relationships inferred bystatistically significant trend tests.” Biometrics. 47, 139–146.

Marcus, R., Peritz, E., Gabriel, K.R. (1976). “On closed testing procedure with special reference toordered analysis of variance.” Biometrika. 63, 655–660.

Mukerjee, H., Robertson, T., Wright, F.T. (1987). “Comparison of several treatments with a controlusing multiple contrasts.” Journal of the American Statistical Association. 82, 902–910.

Neuhauser, M., Liu, P.Y., Hothorn, L.A. (1998). “Nonparametric tests for trend: Jonckheere’s test,a modification and a maximum test.” Biometrical Journal. 40, 899–909.

Phillips, A. (1997). “Design and analysis of dose response studies: reality versus regulatoryrequirements.” Drug Information Journal. 31, 737–744.

Page 324: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 11 Design and Analysis of Dose-Ranging Clinical Studies 311

Phillips, A. (1998). “A review of the performance of tests used to establish whether there is a drugeffect in dose-response studies.” Drug Information Journal. 32, 683–692.

Rom, D.M., Costello, R.J., Connell, L.T. (1994). “On closed test procedures for dose-responseanalysis.” Statistics in Medicine. 13, 1583–1596.

Ruberg, S.J. (1989). “Contrasts for identifying the minimum effective dose.” Journal of theAmerican Statistical Association. 84, 816–822.

Ruberg, S.J. (1995a). “Dose response studies. Some design considerations.” Journal ofBiopharmaceutical Statistics. 5, 1–14.

Ruberg, S.J. (1995b). “Dose response studies. Analysis and interpretation.” Journal ofBiopharmaceutical Statistics. 5, 15–42.

Shirley, E.A. (1977). “A nonparametric equivalent of Williams’ test for contrasting increasing doselevels of a treatment.” Biometrics. 33, 386–389.

Shirley, E.A. (1985). “The value of specialized tests in studies where ordered group means areexpected.” Statistics in Medicine. 4, 489–496.

Stefansson, G., Kim, W.C., Hsu, J.C. (1988). “On confidence sets in multiple comparisons.”Statistical Decision Theory and Related Topics IV. Edited by S.S. Gupta and J.O. Berger. NewYork: Academic Press. 89–102.

Stewart, W.H., Ruberg, S.J. (2000). “Detecting dose response with contrasts.” Statistics inMedicine. 19, 913–921.

Tamhane, A.C., Hochberg, Y., Dunnett, C.W. (1996). “Multiple test procedures for dose finding.”Biometrics. 52, 21–37.

Tamhane, A.C., Logan, B.R. (2002). “Multiple test procedures for identifying the minimumeffective and maximum safe doses of a drug.” Journal of the American Statistical Association.97, 293–301.

Terpstra, T.J. (1952). “The asymptotic normality and consistency of Kendall’s test against trend,when ties are present in one ranking.” Indigationes Mathematicae. 14, 327–333.

Tukey, J.W., Ciminera, J.L., Heyse, J.F. (1985). “Testing the statistical certainty of a response toincreasing doses of a drug.” Biometrics. 41, 295–301.

Westfall, P.H. (1997). “Multiple testing of general contrasts using logical constraint andcorrelations.” Journal of the American Statistical Association. 92, 299–306.

Westfall, P.H., Tobias, R.D. (2006). “Multiple testing of general contrasts: truncated closure andthe extended Shaffer-Royen method.” Journal of the American Statistical Association. In press.

Westfall, P.H., Tobias, R.D., Rom, D., Wolfinger, R.D., Hochberg, Y. (1999). Multiple Comparisonsand Multiple Tests Using the SAS System. Cary, NC: SAS Institute Inc.

Williams, D.A. (1971). “A test for differences between treatment means when several dose levels arecompared with a zero dose control.” Biometrics. 27, 103–117.

Williams, D.A. (1972). “The comparison of several dose levels with a zero dose control.” Biometrics.28, 519–531.

Williams, D.A. (1988). “Tests for differences between several small proportions.” Applied Statistics.37, 421–434.

Page 325: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 326: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12

Analysis of Incomplete Data

Geert MolenberghsCaroline BeunckensHerbert ThijsIvy JansenGeert VerbekeMichael KenwardKristel Van Steen

12.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314

12.2 Case Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316

12.3 Data Setting and Modeling Framework . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 318

12.4 Simple Methods and MCAR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319

12.5 MAR Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320

12.6 Categorical Data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322

12.7 MNAR Modeling. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .340

12.8 Sensitivity Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347

12.9 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356

Relying on Rubin’s standard missing-data taxonomy, and using simple algebraicderivations, this chapter argues that some methods that are commonly used to handleincomplete longitudinal data are based on poor principles and are unnecessarily restrictive.We define longitudinal clinical trial data as complete case analyses and methods based onlast observation carried forward (LOCF), for which the missing completely at random(MCAR) assumption is required.

Because flexible software is available that can analyze longitudinal sequences of unequallength, this chapter proposes a shift to a likelihood-based ignorable analysis that is carriedout using SAS software.

Geert Molenberghs is Professor, Center for Statistics, Universiteit Hasselt, Belgium. Caroline Beunckens is Research As-sistant, Center for Statistics, Universiteit Hasselt, Belgium. Herbert Thijs is Postdoctoral Fellow, Center for Statistics,Universiteit Hasselt, Belgium. Ivy Jansen is Postdoctoral Fellow, Center for Statistics, Universiteit Hasselt, Belgium. GeertVerbeke is Professor, Biostatistical Center, Katholieke Universiteit Leuven, Belgium. Michael Kenward is Professor, Med-ical Statistics Unit, London School of Hygiene and Tropical Medicine, United Kingdom. Kristel Van Steen is Professor,Universitiet Gent, Belgium.

Page 327: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

314 Pharmaceutical Statistics Using SAS: A Practical Guide

12.1 IntroductionIn a longitudinal clinical trial, each unit is measured on several occasions. It is not unusualin practice for some sequences of measurements to terminate early for reasons outside thecontrol of the investigator. Any unit so affected is called a dropout. It might therefore benecessary to accommodate the dropout in the modeling process.

When referring to the missing-value (or non-response) process we will use theterminology of Little and Rubin (1987, Chapter 6). A non-response process is said to bemissing completely at random (MCAR) if the missingness is independent of both unobservedand observed data. A process is said to be missing at random (MAR) if, conditional on theobserved data, the missingness is independent of the unobserved measurements. A processthat is neither MCAR nor MAR is termed non-random (MNAR). In the context oflikelihood inference, and when the parameters describing the measurement process arefunctionally independent of the parameters describing the missingness process, MCAR andMAR are ignorable, while a non-random process is non-ignorable.

Many methods are formulated as selection models (Little and Rubin, 1987) as opposedto pattern-mixture models (PMM) (Little 1993, 1994a). A selection model factors the jointdistribution of the measurement and response mechanisms into the marginal measurementdistribution and the response distribution, conditional on the measurements. This isintuitively appealing since the marginal measurement distribution would be of interest alsowith complete data. Little and Rubin’s taxonomy is most easily developed in the selectionsetting. Parameterizing and making inference about the effect of treatment and itsevolution over time is straightforward in the selection model context.

12.1.1 Incomplete Data in Clinical TrialsIn the specific case of a clinical trial setting, standard methodology used to analyzelongitudinal data subject to non-response is mostly based on such methods as lastobservation carried forward (LOCF), complete case analysis (CC), or simple forms ofimputation. This is often done without questioning the possible influence of theseassumptions on the final results, even though several authors have written about this topic.A relatively early account is given in Heyting, Tolboom, and Essers (1992). Mallinckrodtet al. (2003a,b) and Lavori, Dawson, and Shera (1995) propose direct-likelihood andmultiple-imputation methods, respectively, to deal with incomplete longitudinal data.Siddiqui and Ali (1998) compare direct-likelihood and LOCF methods.

It is unfortunate that there is such a strong emphasis on methods like LOCF and CC,since they are based on extremely strong assumptions. In particular, even the strongMCAR assumption does not suffice to guarantee that an LOCF analysis is valid. On theother hand, under MAR, valid inference can be obtained through a likelihood-basedanalysis, without the need for modeling the dropout process. As a consequence, we cansimply use, for example, linear or generalized linear mixed models (Verbeke andMolenberghs, 2000), without additional complication or effort. We will argue that such ananalysis not only enjoys much wider validity than the simple methods but in addition issimple to conduct, without additional data manipulation using such tools as, for example, theSAS MIXED or NLMIXED procedure. Thus, clinical trial practice should shift away fromthe ad hoc methods and focus on likelihood-based, ignorable analyses instead. As will beargued further, the cost involved in having to specify a model will arguably be mild tomoderate in realistic clinical trial settings. Thus, we promote the use of direct-likelihoodignorable methods and argue against the use of the LOCF and CC approaches.

At the same time, we cannot avoid a reflection on the status of MNAR approaches. Inrealistic settings, the reasons for dropout are varied, and it is therefore difficult to fullyjustify on a priori grounds the assumption of MAR. For example, the rate of and thereasons for dropout varied considerably across eleven clinical trials of similar design, of the

Page 328: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 315

same drug in the same indication. In one study, completion rates were 80% for drug andplacebo. In another study, two-thirds of the patients who were taking a drug completed thestudy, while only one-third did so on placebo. In yet another study, 70% finished onplacebo but only 60% on drug. Reasons for dropout also varied, even within the drug arm.For example, at low doses, more patients on drug dropped out due to lack of efficacy,whereas at higher doses dropout that was due to adverse events was more common. At firstsight, this calls for a further shift towards MNAR models. However, some carefulconsiderations have to be made, the most important one of which is that no modelingapproach, whether either MAR or MNAR, can recover the lack of information that occursdue to incompleteness of the data.

First, under MAR, a standard analysis would follow, if it would be possible to beentirely sure of the MAR nature of the mechanism. However, it is only rarely the case thatsuch an assumption is known to hold (Murray and Findlay, 1988). Nevertheless, ignorableanalyses may provide reasonably stable results, even when the assumption of MAR isviolated, in the sense that such analyses constrain the behavior of the unseen data to besimilar to that of the observed data (Mallinckrodt et al., 2001a,b). A discussion of thisphenomenon in the survey context has been given in Rubin, Stern, and Vehovar (1995).These authors argue that, in well-conducted experiments (some surveys and manyconfirmatory clinical trials), the assumption of MAR is often to be regarded as a realisticone. Second, and very important for confirmatory trials, an MAR analysis can be specifieda priori without additional work relative to a situation with complete data. Third, whileMNAR models are more general and explicitly incorporate the dropout mechanism, theinferences they produce are typically highly dependent on the untestable and often implicitassumptions built in regarding the distribution of the unobserved measurements given theobserved ones. The quality of the fit to the observed data need not reflect at all theappropriateness of the implied structure governing the unobserved data. This point isirrespective of the MNAR route taken, whether a parametric model of the type of Diggleand Kenward (1994) is chosen, or a semiparametric approach such as in Robins, Rotnitzky,and Scharfstein (1998). Hence in any incomplete-data setting there cannot be anythingthat could be termed a definitive analysis. Based on these considerations, we recommend,for primary analysis purposes, the use of ignorable likelihood-based methods. In manyexamples, however, the reasons for dropout will be many and varied. It is therefore difficultto justify on a priori grounds the MAR assumption. Arguably, in the presence of MNARmissingness, a wholly satisfactory analysis of the data is not feasible.

In fact, modeling in this context often rests on strong (untestable) assumptions andrelatively little evidence from the data themselves. Glynn, Laird, and Rubin (1986)indicated that this is typical for selection models. It is somewhat less the case forpattern-mixture models (Little, 1993; 1994a; Hogan and Laird, 1997), although cautionshould be used (Thijs, Molenberghs, and Verbeke, 2000). This awareness and the resultingskepticism about fitting MNAR models initiated the search for methods to investigate theresults with respect to model assumptions and for methods that allow us to assessinfluences in the parameters describing the measurement process, as well as the parametersdescribing the non-random part of the dropout mechanism. Several authors have suggestedvarious types of sensitivity analyses to address this issue (Molenberghs, Kenward, andGoetghebeur, 2001; Scharfstein, Rotnitzky, and Robins, 1999; Van Steen et al., 2001;Verbeke et al., 2001). Verbeke et al. (2001) and Thijs, Molenberghs, and Verbeke (2000)developed a local influence-based approach for the detection of subjects that stronglyinfluence the conclusions. These authors focused on the Diggle and Kenward (1994) modelfor continuous outcomes. Van Steen et al. (2001) adapted these ideas to the model ofMolenberghs, Kenward and Lesaffre (1997), for monotone repeated ordinal data. Jansenet al. (2003) focused on the model family proposed by Baker, Rosenberger, andDerSimonian (1992, henceforth referred to as BRD).

Page 329: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

316 Pharmaceutical Statistics Using SAS: A Practical Guide

Thus, to explore the impact of deviations from the MAR assumption on the conclusions,we should ideally conduct a sensitivity analysis, within which MNAR models can play amajor role, together with, for example, pattern-mixture models (Verbeke and Molenberghs,2000, Chapters 18–20).

12.1.2 OutlineThree case studies used throughout this chapter, the exercise bike data and the mastitis indairy cattle data, are introduced in Section 12.2. The general data setting is introduced inSection 12.3, as well as a formal framework for incomplete longitudinal data. A briefoverview on the problems associated with simple methods is presented in Section 12.4.Next, methods valid under the MAR assumption are described in Sections 12.5 and 12.6,for continuous and categorical outcomes respectively. MNAR modeling is covered inSection 12.7, while Section 12.8 covers the aspects on sensitivity analysis.

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

12.2 Case StudiesWe will present three case studies which will be used throughout this chapter.

EXAMPLE: Exercise trialIn this heart failure study, the primary efficacy endpoint is based upon the ability to dophysical exercise. This ability is measured in the number of seconds a subject is able to ridean exercise bike. The data collected in the study are included in the EXERCISE data setthat can be found on the book’s companion Web site. There are 25 subjects assigned toplacebo (GROUP=0) and 25 to treatment (GROUP=1). The treatment consisted of theadministration of ACE inhibitors. Four measurements were taken at monthly intervals(TIME variable). The Y variable represents the outcome scores transformed to normality.

All 50 subjects are observed at the first occasion, whereas there are 44, 41, and 38subjects seen at the second, the third, and the fourth visits, respectively. Individual andmean response profiles per treatment arm are shown in Figures 12.1 and 12.2, respectively.The percentage of patients remaining in the study after each visit is tabulated inTable 12.1 per treatment arm.

Table 12.1 Percentage of Patients Remaining inthe Study per Treatment Arm in the Exercise Trial

Visit Placebo Treatment

1 100% 100%2 88% 88%3 84% 80%4 80% 72%

EXAMPLE: Mastitis dataThis example, concerning the occurrence of the infectious disease mastitis in dairy cows,was introduced in Diggle and Kenward (1994) and reanalyzed in Kenward (1998). TheMASTITIS data set, provided on the book’s companion Web site, contains the milk yields(in thousands of liters) of 107 dairy cows from a single herd in two consecutive years:Yij(i = 1, . . . , 107; j = 1, 2). In the first year, all animals were supposedly free of mastitis; in

Page 330: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 317

Figure 12.1 Individual response profiles in the placebo (left panel) and treatment (right panel) groups in theexercise trial

Res

pons

e

-5

0

5

10

Visit

1 2 3 4

Res

pons

e

-5

0

5

10

Visit

1 2 3 4

Figure 12.2 Mean response profiles in the placebo (solid curve) and treatment (dashed curve) groups in theexercise trial

Res

pons

e

0

2

4

6

Visit

1 2 3 4

the second year, 27 became infected. Mastitis typically reduces milk yield, and the questionof scientific interest is whether the probability of occurrence of mastitis is related to theyield that would have been observed had mastitis not occurred. A graphical representationof the complete data is given in Figure 12.3.

EXAMPLE: Depression trialThe DEPRESSION data set available on the book’s companion Web site comes from aclinical trial including 342 patients with post-baseline data. The Hamilton DepressionRating Scale (HAMD17) is used to measure the depression status of the patients. For eachpatient, BASVAL represents the baseline HAMD17 assessment, Y is the HAMD17 score atVisits 4 through 8 and CHANGE is the change from baseline in the HAMD17 score atVisits 4 through 8. The YBIN variable is a binary outcome derived from the Y variable,i.e., Y=1 if the HAMD17 score is larger than 7, and 0 otherwise. Two treatment groups

Page 331: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

318 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 12.3 Scatter plots of the Year 2 milk yield versus the Year 1 milk yield (left panel) the change in milkyield versus the Year 1 milk yield (right panel) in the mastitis example. Black dots represent Cows #4 and #5.

Yea

r 2

milk

yie

ld

2

4

6

8

10

12

Year 1 milk yield

2 4 6 8 10 12

Cha

nge

in m

ilk y

ield

-2

0

2

4

6

Year 1 milk yield

2 4 6 8 10 12

(primary dose of experimental drug, TRT=1, and placebo, TRT=4) are included in thedata set.

Mean profiles of the HAMD17 changes and dropout rates in the two treatment groupsare shown in Figure 12.4.

Figure 12.4 Left panel: Mean response profiles in Treatment groups 1 (solid curve) and 4 (dashed curve) inthe depression trial. Right panel: Percentage of patients remaining in the study Treatment groups 1 and 4.

Cha

nge

in H

AM

D17

-11

-9

-7

-5

-3

-1

Visit

4 5 6 7 8

Rem

aini

ng p

atie

nts

(%)

50

60

70

80

90

100

Visit

4 5 6 7 8

12.3 Data Setting and Modeling FrameworkAssume that for subject i = 1, . . . , N in the study a sequence of responses Yij is designed tobe measured at occasions j = 1, . . . , n. The outcomes are grouped into a vectorYi = (Yi1, . . . , Yin)′. In addition, define a dropout indicator Di for the occasion at whichdropout occurs and make the convention that Di = n + 1 for a complete sequence. It isoften necessary to split the vector Yi into observed (Y o

i ) and missing (Y mi ) components

Page 332: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 319

respectively. Note that dropout is a particular case of monotone missingness. In order tohave a monotone pattern of missingness, there has to exist a permutation of themeasurement components such that a measurement earlier in the permuted sequence isobserved for at least those subjects that are observed at later measurements. For thisdefinition to be meaningful, we need to have a balanced design in the sense of a commonset of measurement occasions. Other patterns are called nonmonotone or intermittentmissingness. When intermittent missingness occurs, it is best to use a vector of binaryindicators Ri = (Ri1, . . . , Rin)′ rather than the dropout indicator Di.

In principle, we would like to consider the density of the full data f(yi, di|θ, ψ), wherethe parameter vectors θ and ψ describe the measurement and missingness processes,respectively. Covariates are assumed to be measured but, for notational simplicity, they aresuppressed from notation.

The taxonomy, constructed by Rubin (1976), further developed in Little and Rubin(1987), and informally sketched in Section 12.1, is based on the factorization

f(yi, di|θ, ψ) = f(yi|θ)f(di|yi, ψ), (12.3.1)

where the first factor is the marginal density of the measurement process and the secondone is the density of the missingness process, conditional on the outcomes. Factorization(12.3.1) forms the basis of selection modeling as the second factor corresponds to the(self-)selection of individuals into observed and missing groups. An alternative taxonomycan be built based on so-called pattern-mixture models (Little, 1993; Little, 1994a). Theseare based on the factorization

f(yi, di|θ, ψ) = f(yi|di, θ)f(di|ψ). (12.3.2)

Indeed, (12.3.2) can be seen as a mixture of different populations, characterized by theobserved pattern of missingness.

Rubin (1976) and Little and Rubin (1987) have shown that, under MAR and mildregularity conditions (parameters θ and ψ are functionally independent), likelihood-basedand Bayesian inferences are valid when the missing data mechanism is ignored (see alsoVerbeke and Molenberghs, 2000; Molenberghs and Verbeke, 2005). Practically speaking,the likelihood of interest is then based upon the factor f(yo

i |θ). This is called ignorability.The practical implication is that a software module with likelihood estimation facilities

and with the ability to handle incompletely observed subjects manipulates the correctlikelihood, providing valid parameter estimates and likelihood ratio values. Dmitrienkoet al. (2005, Chapter 5) provide detailed guidelines on how to implement such analyses.They also issue a number of cautionary remarks. An important one is that the flexibilityand ease of MAR (and hence ignorable) analyses do not rule out the option of an MNARmechanism to operate. These authors also focused primarily on continuous outcomes. Inthis chapter, both discrete outcomes, as well as modeling approaches under MNAR, are ofprimary interest.

12.4 Simple Methods and MCARWe will briefly review a number of relatively simple methods that have been and are still inextensive use. They have been discussed in some detail in Dmitrienko et al. (2005,Chapter 5). We will focus on complete case analysis (CC) and last observation carriedforward (LOCF). The latter is a single or simple imputation method, which shares acertain number of pitfalls with the other methods. Multiple imputation, on the other hand(Section 12.5.2) is valid under MAR and therefore is discussed in Section 12.5.

Page 333: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

320 Pharmaceutical Statistics Using SAS: A Practical Guide

Complete Case AnalysisA complete case (CC) analysis considers only those cases for which all ni measurementswere recorded. The method is simple since it restores balance, in the sense that arectangular data matrix is obtained. However, the drawbacks surpass the advantages. Apartfrom considerable information loss, leading to inefficient estimates and tests with less thanoptimal power, often severe bias is to be expected. The method is, therefore, notrecommended. See Dmitrienko et al. (2005, Chapter 5) for details.

Last Observation Carried ForwardAlternatively, balance can be restored by substituting the last obtained measurement forthe missing ones. This technique is termed last observation carried forward (LOCF) or lastvalue carried forward. The practical advantages are the same as with CC, but the issues aremanifold. The technique has been discussed in detail in Dmitrienko et al. (2005, Chapter 5)and insightful illustrations of the issues are provided in Molenberghs et al. (2004). Animportant issue is that filled-in values are treated as actual data. Further, Molenberghset al. (2004) have illustrated that the bias resulting from this method can be bothconservative and liberal, contradicting common belief that the appeal of the method lies init being conservative for the assessment of treatment effect in superiority trials.

Moreover, since direct likelihood is perfectly feasible in the sense that it is valid underMAR and easy to implement in standard software (Section 12.5), there is generally verylittle reason to apply LOCF. See also Mallinckrodt et al. (2003ab).

Available Case Methods

Available case methods (Little and Rubin, 1987) use as much of the data as possible. In amultivariate normal setting, means and variances are estimated based on the informationavailable for individual outcomes, while covariances are estimated using pairs of outcomes.While this is a simple solution in this setting, the method is difficult to extend to othersettings, such as continuous data to which regression models or linear mixed models areapplied, or categorical data. Moreover, the method is still valid only under MCAR. Thereason is that the method uses moments only; in our setting, these are the first and secondmoments of the multivariate normal. As such, it is a frequentist method. In the followingsection, we will show that using the available information in a likelihood frameworkextends validity to MAR.

12.5 MAR MethodsIn this section, we give an overview of the most commonly used methods, which are validunder the MAR assumption. See Dmitrienko et al. (2005, Chapter 5) for details.

12.5.1 Direct Likelihood AnalysesAs stated earlier, likelihood-based inference is valid whenever the mechanism is MAR,provided that the technical condition holds that the parameters describing the nonresponsemechanism are distinct from the measurement model parameters (Little and Rubin, 1987).The log-likelihood then partitions into two functionally independent components, onedescribing the measurement model, the other one the missingness model. This implies thatlikelihood-based software with facilities to handle incomplete records provides validinferences. Such a likelihood-based ignorable analysis is also termed likelihood-based MARanalysis, or, as we will call it further on, a direct likelihood analysis.

Turning to SAS software, this implies that likelihood-based longitudinal analyses,conducted by means of the MIXED, NLMIXED, and GLIMMIX procedures, are valid,

Page 334: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 321

given that the MAR assumption holds or is considered a reasonable approximation. This isuseful, even when inferences are conducted at the end of the planned measurementsequence. An important example is the assessment of treatment effect at the end of aclinical trial. A way to practically implement such an analysis is by specifying a sufficientlygeneral treatment by time-mean profile model, supplemented with an unstructuredvariance-covariance structure. Appropriate use of the CONTRAST or ESTIMATEstatement then leads to the required test. Technically, the score equations take theexpected value of the incomplete measurements, given the observed ones, into account(Beunckens, Molenberghs, and Kenward, 2005). This implies that all information on asubject is used to assess treatment effect. If the treatment assignment is used asrandomized, the method is fully consistent with the intention-to-treat principle.

12.5.2 Multiple ImputationApart from direct likelihood, other methods that are valid under MAR include multipleimputation (Rubin 1978, 1987, Rubin and Schenker 1986) (discussed here) and theExpectation-Maximization algorithm.

The key idea of the multiple imputation (MI) procedure is to replace each missing valuewith a set of M plausible values. Precisely, such values are drawn from the conditionaldistribution of the unobserved values, given the observed ones. The imputed data sets arethen analyzed using standard complete data methods and software. Finally, the Mobtained inferences thus obtained must be combined into a single one by means of themethod proposed by Rubin (1978). Ample detail can be found in Dmitrienko et al. (2005,Chapter 5).

With the availability and ease of direct likelihood, it remains to be discussed when touse multiple imputation. First, the method can be used to conduct checks on directlikelihood. Second, MI is really useful when there are incomplete covariates, along withmissing outcomes. Third, when several users want to conduct a variety of analyses on thesame incomplete set of data, it is sensible to provide all of them with the same multiplyimputed sets of data. Finally, multiple imputation can be used within the context ofsensitivity analysis.

The SAS MI procedure is a multiple imputation procedure that creates multiplyimputed data sets for incomplete p-dimensional multivariate data. It uses methods thatincorporate appropriate variability across the M imputations. Once the M complete datasets are analyzed by using standard procedures, the MIANALYZE procedure can be usedto generate valid statistical inferences about these parameters by combining results for theM complete data sets. See also Dmitrienko et al. (2005, Chapter 5).

12.5.3 The EM AlgorithmThe Expectation-Maximization (EM) algorithm has been described in detail in Dmitrienkoet al. (2005, Chapter 5). It is an alternative to direct likelihood and MI, in the sense that,in its basic form, it is valid under the same conditions. Dempster, Laird, and Rubin (1977)provided a very general description of the algorithm, showing its use in broad classes ofmissing data, latent variable, latent classes, random effects, and other data augmentationsettings.

Within each iteration, there are two steps. In the E step, the expectation of the completedata log-likelihood is calculated. In exponential family settings, this is particularly easysince it reduces to the calculation of complete-data sufficient statistics. In the M step, theso-obtained function is maximized. Little and Rubin (1987), Schafer (1997), and McLachlanand Krishnan (1997) provide detailed descriptions and applications of the EM algorithm.

While the algorithm is elegant, the basic version does not provide precision estimatesand a number of proposals have been made over the years, summarized in McLachlan and

Page 335: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

322 Pharmaceutical Statistics Using SAS: A Practical Guide

Krishnan (1997), to rectify this situation. A number of applications of the EM algorithmare possible with the SAS MI procedure (Dmitrienko et al. 2005, Chapter 5).

12.6 Categorical DataFor non-Gaussian outcomes, there is no single, broadly applicable counterpart to themultivariate normal model, within which the linear mixed model is positioned. Therefore,it is important to carefully distinguish between three model families: marginal models,random-effects models, and conditional models. A comprehensive introduction of these andcomparison between them has been provided in Dmitrienko et al. (2005, Chapter 5). Here,we provide a brief overview of the marginal family, with focus on generalized estimatingequations (GEE), and put some emphasis on the random-effects family. In particular, weemphasize the generalized linear mixed model (GLMM).

12.6.1 Marginal ModelsMarginal models describe the outcomes within an outcome sequence Yi, conditional oncovariates, but neither on other outcomes nor on unobserved (latent) structures. While fulllikelihood approaches exist (Molenberghs and Verbeke 2005), they are usually demandingin computational terms, explaining the popularity of generalized estimating equations(GEE), on which we will focus here. In their basic form, they are valid under MCAR,which explains why weighted GEE (WGEE) have been devised to allow for extension tothe MAR framework. Whereas GEE can be fitted using the GENMOD procedure, therealso is a linearization-based version, which can be fitted with the %GLIMMIX macro orprocedure. A number of additional extensions of, and modifications to, GEE exist. Theyare not considered here. See Molenberghs and Verbeke (2005) for details.

Generalized Estimating Equations

Generalized estimating equations (Liang and Zeger, 1986) are useful when scientific interestfocuses on the first moments of the outcome vector. Examples include time evolutions inthe response probability, treatment effect, their interaction, and the effect of (baseline)covariates on these probabilities. GEE allows the researcher to use a “fix up” for thecorrelations in the second moments, and to ignore the higher order moments, while stillobtaining valid inferences, at reasonable efficiency.

The GEE methodology is based on solving the equations

S(β) =N∑

i=1

∂μi

∂β′ (A1/2i RiA

1/2i )−1(yi − μi) = 0, (12.6.3)

in which the marginal covariance matrix Vi has been decomposed in the form A1/2i RiA

1/2i ,

with Ai the matrix with the marginal variances on the main diagonal and zeros elsewhere,and with Ri = Ri(α) the marginal correlation matrix, often referred to as the workingcorrelation matrix. Usually, the marginal covariance matrix Vi = A

1/2i RiA

1/2i contains a

vector α of unknown parameters which is replaced for practical purposes by a consistentestimate.

Assuming that the marginal mean μi has been correctly specified as h(μi) = Xiβ, it canbe shown that, under mild regularity conditions, the estimator β obtained from solving(12.6.3) is asymptotically normally distributed with mean β and with covariance matrix

I−10 I1I

−10 , (12.6.4)

Page 336: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 323

where

I0 =

(N∑

i=1

∂μi′

∂βV −1

i

∂μi

∂β′

), I1 =

(N∑

i=1

∂μi′

∂βV −1

i Var(yi)V −1i

∂μi

∂β′

).

In practice, Var(yi) in (12.6.4) is replaced by (yi − μi)(yi − μi)′, which is unbiased on thesole condition that the mean was again correctly specified.

Note that valid inferences can now be obtained for the mean structure, only assumingthat the model assumptions with respect to the first-order moments are correct.

Liang and Zeger (1986) proposed moment-based estimates for the working correlation.To this end, first define deviations:

eij =yij − μij√

v(μij)

and decompose the variance slightly more generally as above in the following way:

Vi = φA1/2i RiA

1/2i ,

where φ is an overdispersion parameter.

Weighted Generalized Estimating EquationsAs stated before, GEE is valid under MCAR. To accommodate MAR missingness, Robins,Rotnitzky, and Zhao (1995) proposed a class of weighted estimating equations. The idea ofWGEE is to weight each subject’s measurements in the GEEs by the inverse probabilitythat a subject drops out at that particular measurement occasion. This can be calculated as

νij = P (Di = j) =

⎧⎪⎪⎪⎪⎪⎪⎨⎪⎪⎪⎪⎪⎪⎩

P (Di = j|Di ≥ j) j = 2,

P (Di = j|Di ≥ j)j−1∏k=2

[1 − P (Di = k|Di ≥ k)] j = 3, . . . , ni,

ni∏k=2

[1 − P (Di = k|Di ≥ k)] j = ni + 1.

In the weighted GEE approach, which is proposed to reduce possible bias of β, the scoreequations to be solved when taking into account the correlation structure are:

S(β) =N∑

i=1

1νi

∂μi

∂β′ (A1/2i RiA

1/2i )−1(yi − μi) = 0

or

S(β) =N∑

i=1

n+1∑d=2

I(Di = d)νid

∂μi(d)∂β′ (A1/2

i RiA1/2i )−1(d)(yi(d) − μi(d)) = 0,

where yi(d) and μi(d) are the first d − 1 elements of yi and μi respectively. We define ∂μi

∂β′ (d)

and (A1/2i RiA

1/2i )−1(d) analogously, in line with the definition of Robins, Rotnitzky, and

Zhao (1995).

A Method Based on Linearization

Both versions of GEE studied so far can be seen as deriving from the score equations ofcorresponding likelihood methods. In a sense, GEE result from considering only a

Page 337: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

324 Pharmaceutical Statistics Using SAS: A Practical Guide

subvector of the full vector of scores, corresponding to either the first moments only (theoutcomes themselves), or to the first and second moments (outcomes and cross-productsthereof). On the other hand, they can be seen as an extension of the quasi-likelihoodprinciples, where appropriate modifications are made to the scores to be sufficiently flexibleand “work” at the same time. A classical modification is the inclusion of an overdispersionparameter, while in GEE also (nuisance) correlation parameters are introduced.

An alternative approach consists of linearizing the outcome, in the sense of Nelder andWedderburn (1972), to construct a working variate, to which then weighted least squares isapplied. In other words, iteratively reweighted least squares (IRLS) can be used(McCullagh and Nelder, 1989). Within each step, the approximation produces all elementstypically encountered in a multivariate normal model, and hence corresponding softwaretools can be used. In case our models would contain random effects as well, the core of theIRLS could be approached using linear mixed models tools.

Write the outcome vector in a classical (multivariate) generalized linear models fashion:

yi = μi + εi

where, as usual, μi = E(yi) is the systematic component and εi is the random component,typically following a multinomial distribution. We assume that Var(yi) = Var(εi) = Σi. Themodel is further specified by assuming

ηi = g(μi), ηi = Xiβ,

where ηi is the usual set of linear predictors, g(·) is a vector link function, typically madeup of logit components, Xi is a design matrix and β are the regression parameters.

Estimation proceeds by iteratively solving

N∑i=1

X ′iWiXiβ =

N∑i=1

Wiy∗i , (12.6.5)

where a working variate y∗i has been defined, following from a first-order Taylor series

expansion of ηi around μi:

y∗i = ηi + (yi − μi)F

−1i , Fi =

∂μi

∂ηi

∣∣∣∣β=β

. (12.6.6)

The weights in (12.6.5) are specified as

Wi = F ′i Σ

−1i Fi. (12.6.7)

In these equations β and Σ are evaluated at the current iteration step. Note that in thespecific case of an identity link, ηi = μi, Fi = Ini , and yi = y∗

i , whence a standardmultivariate regression follows.

The linearization based method can be implemented using the %GLIMMIX macro andPROC GLIMMIX, by ensuring no random effects are included. Empirically correctedstandard errors can be obtained by including the EMPIRICAL option.

12.6.2 Random-Effects ModelsModels with subject-specific parameters are differentiated from population-averagedmodels by the inclusion of parameters which are specific to the cluster. Unlike thecorrelated Gaussian outcomes, the parameters of the random effects and population-averaged models for correlated binary data describe different types of effects of thecovariates on the response probabilities (Neuhaus, 1992).

Page 338: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 325

The choice between population-averaged and random-effects strategies should heavilydepend on the scientific goals. Population-averaged models evaluate the overall risk as afunction of covariates. With a subject-specific approach, the response rates are modeled asa function of covariates and parameters, specific to a subject. In such models,interpretation of fixed-effects parameters is conditional on a constant level of therandom-effects parameter. Population-averaged comparisons, on the other hand, make nouse of within-cluster comparisons for cluster-varying covariates and are therefore not usefulto assess within-subject effects (Neuhaus, Kalbfleisch and Hauck, 1991).

Whereas the linear mixed model is unequivocally the most popular choice in the case ofnormally distributed response variables, there are more options in the case of non-normaloutcomes. Stiratelli, Laird, and Ware (1984) assume the parameter vector to be normallydistributed. This idea has been carried further in the work on so-called generalized linearmixed models (Breslow and Clayton, 1993) which is closely related to linear and non-linearmixed models. Alternatively, Skellam (1948) introduced the beta-binomial model, in whichthe response probability of any response of a particular subject comes from a betadistribution. Hence, this model can also be viewed as a random-effects model. We willconsider generalized linear mixed models.

Generalized Linear Mixed ModelsPerhaps the most commonly encountered subject-specific (or random-effects) model is thegeneralized linear mixed model. A general framework for mixed-effects models can beexpressed as follows.

As before, Yij , is the jth outcome measured for cluster (subject) i, i = 1, . . . , N ,j = 1, . . . , ni and Yi is the ni-dimensional vector of all measurements available for cluster i.It is assumed that, conditional on q-dimensional random effects bi, assumed to be drawnindependently from the N(0, D), the outcomes Yij are independent with densities of theform

fi(yij |bi, β, φ) = exp{φ−1[yijθij − ψ(θij)] + c(yij , φ)

},

with η(μij) = η(E(Yij|bi)) = xij′β + zij

′bi for a known link function η(·), with xij and zij

p-dimensional and q-dimensional vectors of known covariate values, with β a p-dimensionalvector of unknown fixed regression coefficients, with φ a scale parameter, and with θij thenatural (or canonical) parameter. Further, let f(bi|G) be the density of the N(0, G)distribution for the random effects bi.

Due to the above independence assumption, this model is often referred to as aconditional independence model. This assumption is the basis of the implementation in theNLMIXED procedure. Just as in the linear mixed model case, the model can be extendedwith residual correlation, in addition to the one induced by the random effects. Such anextended model has been implemented in the SAS GLIMMIX procedure, and itspredecessor the %GLIMMIX macro. This advantage is counterbalanced by bias induced bythe optimization routine employed by GLIMMIX. It is important to realize that GLIMMIXcan be used without random effects as well, thus effectively producing a marginal model,with estimates and standard errors similar to those obtained with GEE.

Note: The GLIMMIX procedure is experimental in SAS 9.1. There are advantages tousing both PROC GLIMMIX and the %GLIMMIX macro, as we shall see.

In general, unless a fully Bayesian approach is followed, inference is based on themarginal model for Yi which is obtained from integrating out the random effects. Thelikelihood contribution of subject i then becomes

fi(yi|β,G, φ) =∫ ni∏

j=1

fij(yij |bi, β, φ) f(bi|G) dbi

Page 339: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

326 Pharmaceutical Statistics Using SAS: A Practical Guide

from which the likelihood for β, D, and φ is derived as

L(β, G, φ) =N∏

i=1

fi(yi|β,G, φ) =N∏

i=1

∫ ni∏j=1

fij(yij |bi, β, φ) f(bi|G) dbi. (12.6.8)

The key problem in maximizing the obtained likelihood is the presence of N integralsover the q-dimensional random effects. In some special cases, these integrals can be workedout analytically. However, since no analytic expressions are available for these integrals,numerical approximations are needed. Here, we will focus on the most frequently usedmethods to do so. In general, the numerical approximations can be subdivided in thosethat are based on the approximation of the integrand, those based on an approximation ofthe data, and those that are based on the approximation of the integral itself. An extensiveoverview of the currently available approximations can be found in Tuerlinckx et al. (2004),Pinheiro and Bates (2000), and Skrondal and Rabe-Hesketh (2004). Finally, in order tosimplify notation, it will be assumed that natural link functions are used, butstraightforward extensions can be applied.

When integrands are approximated, the goal is to obtain a tractable integral such thatclosed-form expressions can be obtained, making the numerical maximization of theapproximated likelihood feasible. Several methods have been proposed, but basically allcome down to Laplace-type approximations of the function to be integrated (Tierney andKadane, 1986).

A second class of approaches is based on a decomposition of the data into the mean andan appropriate error term, with a Taylor series expansion of the mean which is a non-linearfunction of the linear predictor. All methods in this class differ in the order of the Taylorapproximation and/or the point around which the approximation is expanded. Morespecifically, consider the decomposition

Yij = μij + εij = h(xijβ + zijbi) + εij (12.6.9)

in which h(·) equals the inverse link function η−1(·), and where the error terms have theappropriate distribution with variance equal to Var(Yij |bi) = φv(μij) for v(·) the usualvariance function in the exponential family. Note that, with the natural link function,

v(μij) =∂h

∂η(xijβ + zijbi).

Several approximations of the mean μij in (12.6.9) can be considered. One possibility isto consider a linear Taylor expansion of (12.6.9) around current estimates β and bi of thefixed effects and random effects, respectively. This will result in the expression

Yi∗ ≡ W−1

i (Yi − μi) + Xiβ + Zibi ≈ Xiβ + Zibi + ε∗i , (12.6.10)

with Wi equal to the diagonal matrix with diagonal entries equal to v(μij), and for ε∗i equal

to W−1i εi, which still has mean zero. Note that (12.6.10) can be viewed as a linear mixed

model for the pseudo data Yi∗, with fixed effects β, random effects bi, and error terms ε∗

i .This immediately yields an algorithm for fitting the original generalized linear mixed

model. Given starting values for the parameters β, G, and φ in the marginal likelihood,empirical Bayes estimates are calculated for bi, and pseudo data Yi

∗ are computed. Then,the approximate linear mixed model (12.6.10) is fitted, yielding updated estimates for β, G,and φ. These are then used to update the pseudo data and this whole scheme is iterateduntil convergence is reached.

The resulting estimates are called penalized quasi-likelihood estimates (PQL) in theliterature (e.g., Molenberghs and Verbeke 2005), or pseudo-quasi-likelihood in theGLIMMIX documentation because they can be obtained from optimizing a quasi-likelihood

Page 340: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 327

function which involves only first- and second-order conditional moments, augmented witha penalty term on the random effects. The pseudo-likelihood terminology derives from thefact that the estimates are obtained by (restricted) maximum likelihood of thepseudo-response or working variable.

An alternative approximation is very similar to the PQL method, but is based on alinear Taylor expansion of the mean muij in (12.6.9) around the current estimates β for thefixed effects and around bi = 0 for the random effects. The resulting estimates are calledmarginal quasi-likelihood estimates (MQL). See Breslow and Clayton (1993) and Wolfingerand O’Connell (1993) for details. Since the linearizations in the PQL and the MQL methodslead to linear mixed models, the implementation of these procedures is often based onfeeding updated pseudo data into software for the fitting of linear mixed models. However,it should be emphasized that the results from these fittings, which are often reportedintermediately, should be interpreted with great care. For example, reported (log)likelihoodvalues correspond to the assumed normal model for the pseudo data and should not beconfused with (log-)likelihood for the generalized linear mixed model for the actual data athand. Further, fitting of linear mixed models can be based on maximum likelihood (ML) aswell as restricted maximum likelihood (REML) estimation. Hence, within the PQL andMQL frameworks, both methods can be used for the fitting of the linear model to thepseudo data, yielding (slightly) different results. Finally, the quasi-likelihood methodsdiscussed here are very similar to the method of linearization discussed in Section 12.6.1 forfitting generalized estimating equations (GEE). The difference is that here, the correlationbetween repeated measurements is modeled through the inclusion of random effects,conditional on which repeated measures are assumed independent, while, in the GEEapproach, this association is modeled through a marginal working correlation matrix.

Note that, when there are no random effects, both this method and GEE reduce to amarginal model, the difference being in the way the correlation parameters are estimated.In both cases, it is possible to allow for misspecification of the association structure byresorting to empirically corrected standard errors. When this is done, the methods are validunder MCAR. In case we would have confidence in the specified correlation structure,purely model-based inference can be conducted, and hence the methods are valid whenmissing data are MAR.

A third method of numerical approximation is based on the approximation of theintegral itself. Especially in cases where the above two approximation methods fail, thisnumerical integration proves to be very useful. Of course, a wide toolkit of numericalintegration tools, available from the optimization literature, can be applied. Several ofthose have been implemented in various software tools for generalized linear mixed models.A general class of quadrature rules selects a set of abscissas and constructs a weighted sumof function evaluations over those. In the particular context of random-effects models,so-called adaptive quadrature rules can be used (Pinheiro and Bates 1995, 2000), where thenumerical integration is centered around the EB estimates of the random effects, and thenumber of quadrature points is then selected in terms of the desired accuracy.

To illustrate the main ideas, we consider Gaussian and adaptive Gaussian quadrature,designed for the approximation of integrals of the form

∫f(z)φ(z)dz, for an known function

f(z) and for φ(z) the density of the (multivariate) standard normal distribution. We willfirst standardize the random effects such that they get the identity covariance matrix. Letδi be equal to δi = G−1/2bi. We then have that δi is normally distributed with mean 0 andcovariance I. The linear predictor then becomes θij = xij

′β + zij′G1/2δi, so the variance

components in G have been moved to the linear predictor. The likelihood contribution forsubject i then equals

fi(yi|β, G, φ) =∫ ni∏

j=1

fij(yij |bi, β, φ) f(bi|G) dbi. (12.6.11)

Page 341: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

328 Pharmaceutical Statistics Using SAS: A Practical Guide

Obviously, (12.6.11) is of the form∫

f(z)φ(z)dz as required to apply (adaptive) Gaussianquadrature.

In Gaussian quadrature,∫

f(z)φ(z)dz is approximated by the weighted sum∫f(z)φ(z)dz ≈

Q∑q=1

wqf(zq).

Q is the order of the approximation. The higher Q, the more accurate the approximationwill be. Further, the so-called nodes (or quadrature points) zq are solutions to the Qthorder Hermite polynomial, while the wq are well-chosen weights. The nodes zq and weightswq are reported in tables. Alternatively, an algorithm is available for calculating all zq andwq for any value Q (Press et al., 1992).

Figure 12.5 Graphical illustration of Gaussian (left panel) and adaptive Gaussian (right panel) quadrature. Thetriangles indicate the position of the quadrature points, while the rectangles indicate the contribution of eachpoint to the integral.

f(z)

phi(z

)

0.0

0.2

0.4

0.6

0.8

z

0 2 4 6

f(z)

phi(z

)

0.0

0.2

0.4

0.6

0.8

z

0 2 4 6

In the case of univariate integration, the approximation consists of subdividing theintegration region in intervals, and approximating the surface under the integrand by thesum of surfaces of the so-obtained approximating rectangles. An example is given in theleft panel of Figure 12.5, for the case of Q = 10 quadrature points. A similar interpretationis possible for the approximation of multivariate integrals. Note that the figure immediatelyhighlights one of the main disadvantages of (non-adaptive) Gaussian quadrature, i.e., thefact that the quadrature points zq are chosen based on φ(z), independent of the functionf(z) in the integrand. Depending on the support of f(z), the zq will or will not lie in theregion of interest. Indeed, the quadrature points are selected to perform well in casef(z)φ(z) approximately behaves like φ(z), i.e., like a standard normal density function.This will be the case, for example, if f(z) is a polynomial of a sufficiently low order. In ourapplications, however, the function f(z) will take the form of a density from theexponential family, hence an exponential function. It may then be helpful to rescale andshift the quadrature points such that more quadrature points lie in the region of interest.This is shown in the right panel of Figure 12.5, and is called adaptive Gaussian quadrature.

In general, the higher the order Q, the better the approximation will be of the Nintegrals in the likelihood. Typically, adaptive Gaussian quadrature needs (many) fewerquadrature points than classical Gaussian quadrature. On the other hand, adaptiveGaussian quadrature requires for each unit the numerical maximization of a function of the

Page 342: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 329

form ln(f(z)φ(z)) for the calculation of z. This implies that adaptive Gaussian quadratureis much more time consuming.

Since fitting of generalized linear mixed models is based on maximum likelihoodprinciples, inferences for the parameters are readily obtained from classical maximumlikelihood theory.

12.6.3 Marginal versus Random-Effects ModelsNote that there is an important difference with respect to the interpretation of the fixedeffects β. Under the classical linear mixed model (Verbeke and Molenberghs, 2000), wehave that E(Yi) equals Xiβ, such that the fixed effects have a subject-specific as well as apopulation-averaged interpretation. Under non-linear mixed models, however, this does nolonger hold in general. The fixed effects now only reflect the conditional effect of covariates,and the marginal effect is not easily obtained anymore as E(Yi) is given by

E(Yi) =∫

yi

∫fi(yi|bi)g(bi)dbidyi.

However, in a biopharmaceutical context, we are often primarily interested in hypothesistesting, and the random-effects framework can be used to this effect.

Note that both WGEE and GLMM are valid under MAR, with the extra condition thatthe model for weights in WGEE has been specified correctly. Nevertheless, the parameterestimates between both are rather different. This is due to the fact that GEE and WGEEparameters have a marginal interpretation, describing average longitudinal profiles,whereas GLMM parameters describe a longitudinal profile, conditional upon the value ofthe random effects. Let us now provide an overview of the differences between marginaland random-effects models for non-Gaussian outcomes (a detailed discussion can be foundin Molenberghs and Verbeke 2004). The interpretation of the parameters in both types ofmodel (marginal or random-effects) is completely different.

Figure 12.6 Representation of model families and corresponding inference (M stands for marginal and REstands for random-effects). A parameter between quotes indicates that marginal functions but no direct marginalparameters are obtained, since they result from integrating out the random effects from the fitted hierarchicalmodel.

model family↙ ↘

marginal random-effectsmodel model

↓ ↓inference inference↙ ↘ ↙ ↘

likelihood GEE marginal hierarchical↓ ↓ ↓ ↓

βM βM βRE (βRE, bi)↓ ↓

“βM” “βM”

A schematic display is given in Figure 12.6. Depending on the model family (marginalor random-effects), we are led to either marginal or hierarchical inference. It is importantto realize that in the general case the parameter βM resulting from a marginal model is

Page 343: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

330 Pharmaceutical Statistics Using SAS: A Practical Guide

different from the parameter βRE even when the latter is estimated using marginalinference. Some of the confusion surrounding this issue may result from the equality ofthese parameters in the very special linear mixed model case. When a random-effectsmodel is considered, the marginal mean profile can be derived, but it will generally notproduce a simple parametric form. In Figure 12.6 this is indicated by putting thecorresponding parameter between quotation marks.

12.6.4 Analysis of Depression Trial DataLet us now analyze the clinical depression trial, introduced in Section 12.2. The binaryoutcome of interest is the YBIN variable (it is equal to 1 if the HAMD17 score is largerthan 7, and 0 otherwise). The primary null hypothesis has been tested using both GEE andWGEE (with the GENMOD procedure), as well as GLMM with adaptive and non-adaptiveGaussian quadrature (with PROC NLMIXED) in Dmitrienko et al. (2005, Chapter 5).Now, we will test the hypothesis using GEE based on linearization, as well as GLMMbased on the penalized quasi-likelihood method. Both methods can be fitted using theGLIMMIX macro. Beginning with SAS 9.1, there is an experimental GLIMMIX procedure,which can be used as well. We include the fixed categorical effects of treatment, visit, andtreatment-by-visit interaction, as well as the continuous, fixed covariates of baseline scoreand baseline score-by-visit interaction. A random intercept will be included whenconsidering the random-effect models.

%GLIMMIX Macro for the Marginal ModelWe will consider the marginal model:

Yij ∼ Bernoulli(μij), logit(

μij

1 − μij

)= Xiβ, (12.6.12)

where Xi is the design matrix for subject i, containing all covariates and an intercept.Program 12.1 fits Model (12.6.12) with exchangeable working assumptions using the

%GLIMMIX macro. The macro is based on fitting the iterative procedure, outlined inSection 12.6.1. The Generalized Linear Models shell linearizes the outcome and computesthe weights, as in (12.6.6) and (12.6.7). Specific statements that govern this procedure arethe ERROR and LINK statements. We chose the binomial error structure. The logit link isthe default for this option, but we have still chosen to specify it, for clarity. The procedureis based on the MIXED procedure, used to solve iteratively reweighted least squaresequations (12.6.5). Virtually all statements that are available in the MIXED procedure canbe used. They are passed on, in string form, to the macro via the STMTS option. Notethat TYPE=CS, referring to compound symmetry, must be used here. For unstructuredworking assumptions, we use TYPE=UN, for AR(1) this would be TYPE=AR(1), and forindependence assumptions, TYPE=SIMPLE needs to be used. One set of assumptions,those corresponding to the PROC MIXED statement, are passed on via a separatestatement, i.e., the PROCOPT string. Note that we have inserted the EMPIRICAL option,to ensure the empirically corrected standard errors are produced. Omitting theEMPIRICAL option produces the model-based standard errors. We have a choice betweenthe updating methods that are available in the MIXED procedure.

To receive the output that would be produced by the MIXED procedure, we can useOPTIONS=MIXPRINTLAST within the STMTS option. To study the PROC MIXEDoutput at each iteration, we add the MIXPRINTALL option. Arguably, the latter isprimarily useful for debugging purposes.

Page 344: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 331

Program 12.1 %GLIMMIX macro for the linearization-based method in the depression trial example

%glimmix(data=depression, procopt=%str(method=ml empirical),stmts=%str(

class patient visit trt;model ybin = visit trt*visit basval basval*visit / solution;repeated visit / subject=patient type=cs rcorr;),

error=binomial,link=logit);

The typical GLIMMIX output consists of tables copied from the MIXED output, as wellas some additional information. Typical output includes bookkeeping information such asmodel information, dimensions, and number of observations. Since we included the RCORRoption in the REPEATED statement, the fitted correlation matrix of the measurements isgiven, which is to be interpreted as the working correlation matrix. In our case, this is a5 × 5 correlation matrix with off-diagonal elements equal to 0.3317, the exchangeableworking correlation. Output 12.1, the Covariance Parameter Estimates portion of theoutput, must be interpreted with caution, though, for reasons that we will explain.

Output from Program 12.1. Covariance Parameter Estimates

Covariance Parameter Estimates

Cov Parm Subject Estimate

CS PATIENT 0.3140Residual 0.6326

The working correlation is obtained from the usual compound-symmetry equation:

0.31400.3140 + 0.6326

= 0.3317.

In the following output, the residual value is copied as the extra-dispersion parameter:

Output from Program 12.1. GLIMMIX Model Statistics

GLIMMIX Model Statistics

Description Value

Deviance 746.6479Scaled Deviance 1180.3240Pearson Chi-Square 673.9938Scaled Pearson Chi-Square 1065.4703Extra-Dispersion Scale 0.6326

It is best not to use this portion of output, since it is not appropriately adapted to thecombination of generalized linear model and the repeated measures nature of the data. Incase we had used independence working assumptions, there would have been a singlecovariance parameter only, which could then be considered the overdispersion parameter.Arguably, there is little basis to do so, and it is unlikely to see almost no overdispersionwith independence and strong underdispersion with exchangeability. It might make moresense to consider the total variance in the exchangeable case, i.e., 0.3140 + 0.6326, the

Page 345: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

332 Pharmaceutical Statistics Using SAS: A Practical Guide

overdispersion parameter. Similarly, it would be better to ignore the fit statistics portion ofoutput, copied from the MIXED procedure.

The most relevant portion of the output is the Solution for Fixed Effects table, with itsassociated F tests.

Output from Program 12.1. Solution and Type 3 Tests for Fixed Effects

Solution for Fixed Effects

Visit StandardEffect Number TRT Estimate Error DF t Value Pr > |t|

Intercept -1.2326 0.7853 168 -1.57 0.1184VISIT 4 0.4457 1.2147 517 0.37 0.7138...VISIT*TRT 8 1 -0.6942 0.3810 517 -1.82 0.0690...BASVAL*VISIT 7 0.02439 0.05170 517 0.47 0.6373BASVAL*VISIT 8 0 . . . .

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

VISIT 4 517 0.39 0.8128VISIT*TRT 5 517 1.22 0.3000BASVAL 1 168 22.17 <.0001BASVAL*VISIT 4 517 1.79 0.1291

These estimates, with both model-based and empirically corrected standard errors, aregiven in the linearization column of Table 12.2, next to the estimates for GEE and WGEE.Clearly, the estimates under exchangeability for the linearization-based method are verysimilar to those for GEE. Since we did not include the treatment as a main effect, theoutput immediately produces the treatment effects at the different time points. Forinstance, the estimate of the effect of treatment at the endpoint is −0.6942, with acorresponding p value equal to 0.0690. Comparing this with the result of GEE (the p-valueof treatment effect at the last visit is 0.0633), it is clear that both are very similar.However, when a WGEE analysis is performed, the result of this contrast changes a lot(p-value is 0.0325).

PROC GLIMMIX for the Marginal ModelWe can also use PROC GLIMMIX to fit models of a marginal type, subject-specific type,as well as models with subject-specific effects and residual association in addition to that.Program 12.2 relies on PROC GLIMMIX to fit a model identical to the model fitted byProgram 12.1. Even though Program 12.2 is rather different, at first sight, fromProgram 12.1, the correspondence is almost immediate. Again, users of PROC MIXED forlinear mixed models will recognize that the code here is very similar to that used in PROCMIXED. The reason is that, internally, PROC GLIMMIX calls PROC MIXED each time alinear mixed model needs to be fitted to newly updated pseudo data.

Page 346: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 333

Table 12.2 Results of Marginal Models in the Depression Trial ExampleNote: Parameter estimates (model-based standard errors; empirically corrected standard errors) for GEE,WGEE and the linearization based method are shown. Main visit effects and interaction terms of baseline andvisit are not shown.

GEE WGEE Linearization

intercept −1.22 (0.77;0.79) −0.70 (0.64;0.86) −1.23 (0.75;0.79)visit 4 0.43 (1.05;1.22) −0.08 (0.88;1.85) 0.45 (1.05;1.22)visit 5 −0.48 (0.91;1.23) −0.13 (0.70;1.55) −0.47 (0.92;1.23)visit 6 0.06 (0.86;1.03) 0.19 (0.72;1.14) 0.05 (0.86;1.03)visit 7 −0.25 (0.89;0.91) −0.28 (0.82;0.88) −0.25 (0.89;0.91)trt × visit 4 −0.24 (0.54;0.57) −1.57 (0.41;0.99) −0.22 (0.53;0.56)trt × visit 5 −0.09 (0.39;0.40) −0.67 (0.21;0.65) −0.08 (0.38;0.40)trt × visit 6 0.17 (0.34;0.35) 0.62 (0.23;0.56) 0.18 (0.34;0.35)trt × visit 7 −0.43 (0.36;0.35) 0.57 (0.30;0.37) −0.42 (0.35;0.35)trt × visit 8 −0.71 (0.38;0.38) −0.84 (0.32;0.39) −0.69 (0.37;0.38)baseline 0.08 (0.04;0.04) 0.07 (0.03;0.05) 0.08 (0.04;0.04)baseline × visit 4 0.12 (0.07;0.07) 0.24 (0.06;0.13) 0.12 (0.06;0.07)baseline × visit 5 0.09 (0.05;0.07) 0.07 (0.04;0.08) 0.09 (0.05;0.07)baseline × visit 6 0.01 (0.05;0.05) 0.01 (0.04;0.06) 0.01 (0.05;0.88)baseline × visit 7 0.02 (0.05;0.05) 0.03 (0.05;0.05) 0.02 (0.05;0.05)

Program 12.2 PROC GLIMMIX for the linearization-based method in the depression trial example

proc glimmix data=depression method=rspl empirical;class patient visit trt;model ybin (event=’1’)=visit trt*visit basval basval*visit/dist=binary solution;random _residual_/subject=patient type=cs;run;

The most important option in PROC GLIMMIX is METHOD. In Program 12.2,METHOD=RSPL specifies the PQL method based on REML for linear mixed models.Note that, strictly speaking, the “penalized” part of PQL is absent, since there are norandom effects and hence no random-effects penalty. In this case, it is more straightforwardto think of restricted pseudo-likelihood (RPL). An overview of the other available optionsis given in Table 12.3.

Table 12.3 Available Options for Specification of the Estimation Method in PROC GLIMMIX

Quasi-likelihood type Inference pseudo-dataGLIMMIX option PQL/MQL ML/REML

METHOD=RSPL PQL REMLMETHOD=MSPL PQL MLMETHOD=RMPL MQL REMLMETHOD=MMPL MQL ML

The CLASS statement in PROC GLIMMIX specifies which variables should beconsidered as factors. Such classification variables can be either character or numeric.Internally, each of these factors will correspond to a set of dummy variables.

The MODEL statement names the response variable and all fixed effects, whichdetermine the Xi matrices. By default, an intercept is added. The EVENT option(EVENT=‘1’) specifies that the probability to be modeled is P (Yij = 1) (the probability of

Page 347: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

334 Pharmaceutical Statistics Using SAS: A Practical Guide

a severe infection) rather than P (Yij = 0). The SOLUTION option is used to request theprinting of the estimates for all the fixed effects in the model, together with standarderrors, t-statistics, corresponding p-values, and confidence intervals. The DIST option isused to specify the conditional distribution of the data, given the random effects. Variousdistributions are available, including the normal, Bernoulli, Binomial, and Poissondistribution. In Program 12.2, the Bernoulli distribution is specified (DIST=BINARY).The default link function is the natural link (in this example, it is the logit link).

The REPEATED statement of the %GLIMMIX macro corresponds to theRANDOM= residual statement in PROC GLIMMIX. SAS refers to this as the R-side ofthe random statement. It is useful to think of it as the variance-covariance matrix of theoutcome vector Yi, of which the variances follow from the mean-variance link, but thecorrelation structure needs to be specified (as in GEE). When random effects are present,this structure refers to the residual correlation, in addition to the correlation induced bythe random effects. Changing the TYPE option in the RANDOM statement to SIMPLE,CS or UN, respectively, combined with either omission or inclusion of the EMPIRICALoption in PROC GLIMMIX produces exactly the same results as with the %GLIMMIXmacro; i.e., the results reported in the third panel of Table 12.2.

The output form PROC GLIMMIX in Program 12.2, although structured differentlyfrom the %GLIMMIX macro output, is largely equivalent. The fact that the empiricallycorrected standard errors are produced is acknowledged:

Output of Program 12.2. Part of the Model Information

The GLIMMIX Procedure

Model Information

Fixed Effects SE Adjustment Sandwich - Classical

The fact that a marginal model (i.e., a model without random effects) is used isacknowledged through reference to the so-called R-side covariance parameters:

Output of Program 12.2. Part of the Dimensions

Dimensions

R-side Cov. Parameters 2

PROC GLIMMIX in Program 12.2 took 11 iterations to converge. The final covarianceparameters are equivalent to the ones produced by the %GLIMMIX macro:

Output of Program 12.2. Covariance Parameter Estimates

Covariance Parameter Estimates

StandardCov Parm Subject Estimate Error

CS PATIENT 0.3196 0.05209Residual 0.6469 0.03955

PROC GLIMMIX also produces the fixed-effects parameters (again equivalent, up tonumerical accuracy, to their %GLIMMIX macro counterparts) together with associated Ftests.

Page 348: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 335

Output from Program 12.2. Solution and Type 3 Tests for Fixed Effects

Solutions for Fixed Effects

Visit StandardEffect Number TRT Estimate Error DF t Value Pr > |t|

Intercept -1.2331 0.7852 168 -1.57 0.1182VISIT 4 0.4463 1.2146 517 0.37 0.7134...VISIT*TRT 8 1 -0.6939 0.3810 517 -1.82 0.0691...BASVAL*VISIT 7 0.02440 0.05170 517 0.47 0.6372BASVAL*VISIT 8 0 . . . .

Type III Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

VISIT 4 517 0.39 0.8129VISIT*TRT 5 517 1.22 0.3003BASVAL 1 168 22.18 <.0001BASVAL*VISIT 4 517 1.79 0.1293

Again as long as PROC GLIMMIX is experimental (as in SAS 9.1), it is a good idea touse the %GLIMMIX macro as well. Also, having the macro as a backup may increase aprogram’s chances of reaching convergence.

PROC GLIMMIX for the Random-Effects Model

PROC GLIMMIX supports the marginal and penalized quasi-likelihood methods. We willfit the following random-effects model, using the PQL method: assume that, conditional onsubject-specific random intercepts, bi, Yij is Bernoulli distributed with mean πij , modeled as

logit(πij) = X iβ + Zibi, (12.6.13)

in which Xi is the design matrix with covariates described above, β is the vector of fixedeffects parameters, Zi is a design matrix for the random effects (in this case a row of ones),and bi is the random intercept assumed to be normally distributed with mean 0 andvariance d.

The procedure has many more statements and options than those presented here, but werestrict our example to the basic statements needed to fit a generalized linear mixed model.

Program 12.3 fits this random-effects model using PQL based on REML estimation forthe linear mixed models for the pseudo data.

Program 12.3 PROC GLIMMIX for the random-effects model using PQL maximization in the depression trialexample

proc glimmix data=depression method=rspl;class patient visit trt;model ybin (event=’1’)=visit trt*visit basval basval*visit/dist=binary solution;random intercept/subject=patient;run;

Page 349: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

336 Pharmaceutical Statistics Using SAS: A Practical Guide

Most of the statements used in Program 12.3 were described above, except for theRANDOM statement. The RANDOM statement defines the random effects in the model,i.e., the Zi matrices containing the covariates with subject-specific regression coefficients.Note that when random intercepts are required (as in this example), this requirementshould be specified explicitly, which is in contrast to the MODEL statement where anintercept is included by default. The SUBJECT option identifies the subjects in theDEPRESSION data set. The variable in the SUBJECT option can be continuous orcategorical (specified in the CLASS statement); however, when it is continuous, PROCGLIMMIX considers a record to be from a new subject whenever the value of this variableis different from the previous record.

Suppose that random slopes for the time trend were to be included as well. This can beachieved by replacing the RANDOM statement in Program 12.3:

random intercept time/subject=patient type=un;run;

Here TYPE=UN specifies that the random-effects covariance matrix G is a generalunstructured 2 × 2 matrix. Special structures are available, e.g., equal variance for theintercepts and slopes, or independent intercepts and slopes.

The output of Program 12.3 includes information about the fitted model and theestimation procedure. The Residual PL estimation technique refers to PQL with REML(restricted or residual maximum likelihood) for the fitting of the linear models for thepseudo data:

Output of Program 12.3. Model Information

The GLIMMIX Procedure

Model Information

Data Set WORK.DEPRESSIONResponse Variable ybinResponse Distribution BinaryLink Function LogitVariance Function DefaultVariance Matrix Blocked By PATIENTEstimation Technique Residual PLDegrees of Freedom Method Containment

The Fit Statistics portion of the output gives minus twice the residuallog-pseudo-likelihood value evaluated in the final solution, together with the informationcriteria of Akaike (AIC) and Schwarz (BIC), as well as a finite-sample corrected version ofAIC (AICC). When REML estimation is used for the fitting of the linear mixed models forthe pseudo data, an objective function is maximized which is called residual log-likelihoodfunction, while, strictly speaking, the function is not a log-likelihood, and should not beused as a log-likelihood. We refer to Verbeke and Molenberghs (2000, Chapters 5 and 6) fora more detailed discussion with examples. Further, information criteria are statistics thatare sometimes used to compare non-nested models which cannot be compared based on aformal testing procedure. The main idea behind information criteria is to compare modelsbased on their maximized (residual) log-likelihood value (or equivalently minimized minustwice the log-likelihood value), while at the same time penalizing for including an excessivenumber of parameters. They should by no means be interpreted as formal statistical testsof significance. In specific examples, different information criteria can even lead to differentmodel selections. An example of this is given in Section 6.4 of Verbeke and Molenberghs

Page 350: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 337

(2000) in the context of linear mixed models. More details about the use of informationcriteria can be found in Akaike (1974), Schwarz (1978), and Burnham and Anderson (1998).

Output of Program 12.3. Fit Statistics and Covariance Parameter Estimates

Fit Statistics

-2 Res Log Pseudo-Likelihood 3488.65Pseudo-AIC (smaller is better) 3490.65Pseudo-AICC (smaller is better) 3490.65Pseudo-BIC (smaller is better) 3493.78Pseudo-CAIC (smaller is better) 3494.78Pseudo-HQIC (smaller is better) 3491.92Pearson Chi-Square 400.28Pearson Chi-Square / DF 0.58

Covariance Parameter Estimates

StandardCov Parm Subject Estimate Error

Intercept PATIENT 2.5318 0.5343

Next, Output 12.3 presents two tables containing estimates and inferences for the fixedeffects in the model. The reported inferences immediately result from the linear mixedmodel fitted to the pseudo data in the last step of the iterative estimation procedure. Fromthe output, it is immediately clear the treatment effect at the last visit is clearlyinsignificant (p-value is 0.1286). Comparing this with the p-value obtained after fittingGLMM with adaptive Gaussian quadrature (p = 0.0954), we see that both yield aninsignificant effect.

Output of Program 12.3. Solutions and Type 3 Tests of Fixed Effects

Solutions for Fixed Effects

Visit StandardEffect Number TRT Estimate Error DF t Value Pr > |t|

Intercept -1.7036 1.0607 167 -1.61 0.1101...VISIT*TRT 8 1 -0.8426 0.5537 518 -1.52 0.1286...BASVAL*VISIT 7 0.02743 0.06897 518 0.40 0.6911BASVAL*VISIT 8 0 . . . .

Type III Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

VISIT 4 518 0.27 0.8959VISIT*TRT 5 518 0.85 0.5141BASVAL 1 518 18.12 <.0001BASVAL*VISIT 4 518 1.20 0.3115

Page 351: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

338 Pharmaceutical Statistics Using SAS: A Practical Guide

The Covariance Parameter Estimates portion of the output lists estimates andassociated standard errors for the variance components in the model, i.e., for the elementsin the random-effects covariance matrix G. In our example, this is the random-interceptsvariance g.

Table 12.4 compares the parameter estimates and standard errors with the resultsobtained by fitting GLMM with adaptive Gaussian quadrature, using PROC NLMIXED.Obviously, there are some differences between these two methods for certain parameters.Since both approaches are likelihood-based, validity under the MAR assumption should befulfilled. For PROC NLMIXED, the integral is approximated using adaptive Gaussianquadrature, which is quite an accurate method, if a sufficient number of quadrature pointsare chosen. However, PROC GLIMMIX (or %GLIMMIX macro) is based on penalizedquasi-likelihood, and thus the mean is approximated by a Taylor expansion. Thisapproximation can be relatively poor for random-effects models and hence the resultsthereof should be treated with caution.

Table 12.4 Results of Random-Effects Models in the Depression Trial ExampleNote: Parameter estimates (standard errors) for GLMM with adaptive Gaussian quadrature (AGQ) andpenalized-quasi-likelihood methods (PQL).

PQL AGQ

intercept −1.70 (1.06) −2.31 (1.34)visit 4 0.66 (1.48) 0.64 (1.75)visit 5 −0.44 (1.29) −0.78 (1.51)visit 6 0.17 (1.22) 0.19 (1.41)visit 7 −0.23 (1.25) −0.27 (1.43)treatment × visit 4 −0.29 (0.66) −0.54 (0.82)treatment × visit 5 −0.10 (0.53) −0.20 (0.68)treatment × visit 6 0.33 (0.49) 0.41 (0.64)treatment × visit 7 −0.47 (0.52) −0.68 (0.67)treatment × visit 8 −0.84 (0.55) −1.20 (0.72)baseline 0.10 (0.06) 0.15 (0.07)baseline × visit 4 0.14 (0.09) 0.21 (0.11)baseline × visit 5 0.12 (0.07) 0.17 (0.09)baseline × visit 6 0.007 (0.07) 0.01 (0.08)baseline × visit 7 0.03 (0.07) 0.03 (0.08)

GLIMMIX Macro for the Random-Effects ModelAlthough still experimental in SAS 9.1, PROC GLIMMIX can be viewed as a formalprocedure that has grown out of the %GLIMMIX macro, applied earlier for fittinggeneralized estimating equations (GEE) based on linearization. In GEE, the associationbetween repeated measures is modeled through a marginal working correlation matrix.Now, this correlation is modeled via the inclusion of random effects, conditional on whichrepeated measures are assumed to be independent. This similarity implies that the samemacro can be used for fitting generalized linear mixed models as well. Without going intomuch detail, we present in Program 12.4 the SAS code needed to repeat the previousanalysis with the %GLIMMIX macro.

Page 352: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 339

Program 12.4 %GLIMMIX macro for the random-effects model using PQL maximization in the depressiontrial example

%glimmix(data=depression,stmts=%str(

class class patient visit trt;model ybin = visit trt*visit basval basval*visit/solution;random intercept/subject=patient;parms (4) (1)/hold=2;),

error=binomial);

The statements that appear in the STMTS statement are directly fed into the PROCMIXED calls needed for fitting the linear mixed models to the pseudo data. Note that bydefault the %GLIMMIX macro includes a residual overdispersion parameter. If thecorresponding generalized linear mixed model does not contain such a parameter, it shouldexplicitly be kept equal to one. This is done using the HOLD option in the PARMSstatement.

Since PROC MIXED uses REML estimation by default, Program 12.4 requests PQLestimation based on REML fitting for the pseudo data. If ML fitting is required, this canbe specified by adding the line

procopt=%str(method=ml),

to the GLIMMIX call in Program 12.4. If MQL is required, rather than the default PQL,this can be specified by adding the following line

options=MQL,

Without discussing the output from the %GLIMMIX macro in much detail, we herepresent some output tables, which can be compared with the output from PROCGLIMMIX (Program 12.3).

Partial Output of Program 12.4

Covariance Parameter Estimates

Cov Parm Subject Estimate

Intercept PATIENT 2.5318Residual 1.0000

Fit Statistics

-2 Res Log Likelihood 3488.6AIC (smaller is better) 3490.6AICC (smaller is better) 3490.7BIC (smaller is better) 3493.8

Page 353: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

340 Pharmaceutical Statistics Using SAS: A Practical Guide

Solution for Fixed Effects

Visit StandardEffect Number TRT Estimate Error DF t Value Pr > |t|

Intercept -1.7036 1.0607 167 -1.61 0.1101...VISIT*TRT 8 1 -0.8426 0.5537 518 -1.52 0.1286...BASVAL*VISIT 7 0.02743 0.06897 518 0.40 0.6911BASVAL*VISIT 8 0 . . . .

Type 3 Tests of Fixed Effects

Num DenEffect DF DF F Value Pr > F

VISIT 4 518 0.27 0.8959VISIT*TRT 5 518 0.85 0.5141BASVAL 1 518 18.12 <.0001BASVAL*VISIT 4 518 1.20 0.3115

Output 12.4 shows that, indeed, the residual overdispersion parameter was set to one.The obtained results are identical to the results produced by PROC GLIMMIX inProgram 12.3.

12.7 MNAR ModelingEven though the assumption of likelihood ignorability encompasses the MAR and not onlythe more stringent and often implausible MCAR mechanisms, it is difficult to exclude theoption of a more general MNAR mechanism. One solution for continuous outcomes is to fitan MNAR model as proposed by Diggle and Kenward (1994). In the discrete case,Molenberghs, Kenward, and Lesaffre (1997) considered a global odds ratio (Dale) model.

However, as pointed out in the introduction and by several authors (Diggle andKenward, 1994; Verbeke and Molenberghs 2000, Chapter 18), we must be extremely carefulwith interpreting evidence for or against MNAR in a selection model context, especially inlarge studies with a lot of power. We will return to these issues in Section 12.8.

12.7.1 Diggle-Kenward ModelTo be consistent with notation introduced in Section 12.3, we assume a vector of outcomesYi is designed to be measured. If dropout occurs, Yi is only partially observed. We denotethe occasion at which dropout occurs by Di > 1, and Yi is split into the(Di − 1)-dimensional observed component Yi

o and the (ni − Di + 1)-dimensional missingcomponent Yi

m. In case of no dropout, we let Di = ni + 1, and Yi equals Yio. The likelihood

contribution of the ith subject, based on the observed data (yio, di), is proportional to the

marginal density function

f(yio, di|θ, ψ) =

∫f(yi, di|θ, ψ) dyi

m =∫

f(yi|θ)f(di|yi, ψ) dyim, (12.7.14)

in which a marginal model for Yi is combined with a model for the dropout process,conditional on the response, and where θ and ψ are vectors of unknown parameters in themeasurement model and dropout model, respectively.

Let hij = (yi1, . . . , yi;j−1) denote the observed history of subject i up to time ti,j−1. TheDiggle-Kenward model for the dropout process allows the conditional probability for

Page 354: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 341

dropout at occasion j, given that the subject was still observed at the previous occasion, todepend on the history hij and the possibly unobserved current outcome yij , but not onfuture outcomes yik, k > j. These conditional probabilities P (Di = j|Di ≥ j, hij , yij , ψ) cannow be used to calculate the probability of dropout at each occasion:

P (Di = j|yi, ψ) = P (Di = j|hij , yij , ψ)

=

⎧⎪⎪⎪⎪⎪⎪⎨⎪⎪⎪⎪⎪⎪⎩

P (Di = j|Di ≥ j, hij , yij , ψ) j = 2,

P (Di = j|Di ≥ j, hij , yij , ψ) ×j−1∏k=2

[1 − P (Di = k|Di ≥ k, hik, yik, ψ)] j = 3, . . . , ni,

ni∏k=2

[1 − P (Di = k|Di ≥ k, hik, yik, ψ)] j = ni + 1.

Diggle and Kenward (1994) combine a multivariate normal model for the measurementprocess with a logistic regression model for the dropout process. More specifically, themeasurement model assumes that the vector Yi of repeated measurements for the ithsubject satisfies the linear regression model Yi ∼ N(Xiβ, Vi), i = 1, . . . , N . The matrix Vi

can be left unstructured or assumed to be of a specific form, e.g., resulting from a linearmixed model, a factor-analytic structure, or spatial covariance structure (Verbeke andMolenberghs, 2000).

In the particular case that a linear mixed model is assumed, we write (Verbeke andMolenberghs, 2000)

Y i = Xiβ + Zibi + εi, (12.7.15)

where Y i is the n dimensional response vector for subject i, 1 ≤ i ≤ N , N is the number ofsubjects, Xi, and Zi are (n × p) and (n × q) known design matrices, β is the p dimensionalvector containing the fixed effects, bi ∼ N(0, G) is the q dimensional vector containing therandom effects. The residual components εi ∼ N(0,Σi).

The logistic dropout model can, for example, take the form

logit [P (Di = j | Di ≥ j, hij , yij , ψ)] = ψ0 + ψ1yi,j−1 + ψ2yij . (12.7.16)

More general models can easily be constructed by including the complete historyhij = (yi1, . . . , yi;j−1), as well as external covariates, in the above conditional dropout model.Note also that, strictly speaking, we could allow dropout at a specific occasion to berelated to all future responses as well. However, this is rather counter-intuitive in manycases. Moreover, including future outcomes seriously complicates the calculations sincecomputation of the likelihood (12.7.14) then requires evaluation of a possiblyhigh-dimensional integral. Note also that special cases of model (12.7.16) are obtained fromsetting ψ2 = 0 or ψ1 = ψ2 = 0, respectively. In the first case, dropout is no longer allowed todepend on the current measurement, implying MAR. In the second case, dropout isindependent of the outcome, which corresponds to MCAR.

Diggle and Kenward (1994) obtained parameter and precision estimates by maximumlikelihood. The likelihood involves marginalization over the unobserved outcomes Yi

m.Practically, this involves relatively tedious and computationally demanding forms ofnumerical integration. This, combined with likelihood surfaces tending to be rather flat,makes the model difficult to use. These issues are related to the problems to be discussednext.

Apart from the technical difficulties encountered during parameter estimation, there arefurther important issues surrounding MNAR based models. Even when the measurementmodel (e.g., the multivariate normal model) would beyond any doubt be the choice ofpreference for describing the measurement process should the data be complete, then the

Page 355: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

342 Pharmaceutical Statistics Using SAS: A Practical Guide

analysis of the actually observed, incomplete version is, in addition, subject to furtheruntestable modeling assumptions.

When missingness is MAR, the problems are less complex, since it has been shown that,in a likelihood or Bayesian framework, it is sufficient to analyze the observed data, withoutexplicitly modeling the dropout process (Rubin 1976, Verbeke and Molenberghs 2000).However, the very assumption of MAR is itself untestable. Therefore, ignoring MNARmodels is as little an option as blindly shifting to one particular MNAR model. A sensiblecompromise between considering a single MNAR model on the one hand or excluding suchmodels from consideration on the other hand, is to study the nature of such sensitivitiesand, building on this knowledge, formulate ways for conducting sensitivity analyses.Indeed, a strong conclusion, arising from most sensitivity analysis work, is that MNARmodels have to be approached cautiously. This was made clear by several discussants to theoriginal paper by Diggle and Kenward (1994), in particular by Laird, Little, and Rubin. Animplication is that, for example, formal tests for the null hypothesis of MAR versus thealternative of MNAR should be approached with the utmost caution, a topic studied indetail by Jansen et al. (2005). These topics are taken up further in Section 12.8.

12.7.2 Implementation of Selection Models in SASWe have developed a series of SAS programs to implement a special case of theDiggle-Kenward approach. For the measurement process, we considered a more specificcase of model (12.7.15), with various fixed effects, a random intercept, and allowingGaussian serial correlation. This means the covariance matrix V i becomes

V i = dJn + σ2In + τ 2Hi, (12.7.17)

where Jn is an n × n matrix with all its elements equal to 1, In is the n × n identity matrix,and Hi is determined through the autocorrelation function ρujk , with ujk the Euclideandistance between tij and tik, i.e.,

Hi =

⎛⎜⎜⎜⎜⎜⎝1 ρu12 · · · ρu1n

ρu12 1 · · · ρu2n

......

. . ....

ρu1n ρu2n · · · 1

⎞⎟⎟⎟⎟⎟⎠ .

The proposed SAS implementation can easily be adapted for another form of model(12.7.15), by just changing this V i matrix.

Program 12.7, available on the book’s companion Web site, plays the central role infitting the Diggle-Kenward model. The following arguments need to be provided to theprogram:

• X and Z matrices that contain all X i and Zi design matrices (i = 1, . . . , N).• Y vector of all Yi response vectors (i = 1, . . . , N).• INITIAL is a vector of initial values for the parameters in the model.• NSUB and NTIME are the number of subjects N and the number of time points n

respectively.

Within Program 12.7, the INTEGR module calculates the integral over the missingdata, and the LOGLIK module evaluates the log-likelihood function L(θ, ψ). Finally, thelog-likelihood function is maximized. Diggle and Kenward (1994) used the simplexalgorithm (Nelder and Mead, 1965) for this purpose and Program 12.7 relies on theNewton-Raphson ridge optimization method (NLPNRR module of SAS/IML) since it

Page 356: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 343

combines stability and speed. However, in other analyses, it may be necessary to try severalother optimization methods available in SAS/IML.

The following NLPNRR call is used in the program:

call nlpnrr(rc,xr,"loglik",initial,opt,con);

Here, LOGLIK is the module of the function we want to maximize. The initial values tostart the optimization method are included in the INITIAL vector. The OPT argumentindicates an options vector that specifies details of the optimization process: OPT[1]=1request maximization, and the amount of output is controlled by OPT[2]. A constraintmatrix is specified in CON, defining lower and upper bounds for the parameters in the firsttwo rows (d > 0, τ 2 > 0 and σ2 > 0). Finally, all optimization methods return the followingresults: the scalar return code, RC, and a row vector, XR. The return code indicates thereason for the termination of the optimization process. A positive return code indicatessuccessful termination, whereas a negative one indicates unsuccessful termination. That is,the result in the XR vector is unreliable. The XR vector contains the optimal point whenthe return code is positive.

Next, the program also calls the NLPFDD module, which is a subroutine thatapproximates derivatives by finite differences method,

call nlpfdd(maxlik,grad,hessian,"loglik",est);

Here, again LOGLIK is the module of the log-likelihood function. The vector that definesthe point at which the functions and derivatives should be computed is EST. This modulecomputes the function values MAXLIK (which is in this case the maximum likelihood,since EST is the maximum likelihood estimate) the gradient vector GRAD, and theHessian matrix HESSIAN, which is needed to calculate the information matrix, and thusthe standard errors STDE.

Analysis of Exercise Study Data

The program for fitting the Diggle-Kenward model is now applied to the analysis of theexercise study data. We will fit the model under the three different missingnessmechanisms, MCAR, MAR, and MNAR. Further, we will also expand the logisticregression for the dropout model by allowing it to depend on covariates.

To obtain initial values for the parameters of the measurement model, Program 12.5 fitsa linear mixed model to the exercise data using PROC MIXED. We assume a linear trendwithin each treatment group, which implies that each profile can be described with twoparameters (intercept and slope). The error matrix is chosen to be of the form (12.7.17).For subject i = 1, . . . , 50 on time point j = 1, . . . , 4, the model can be expressed as

Yij = β0 + β1 (groupi − 1) + β2 tj (groupi − 1) + β3 tj groupi + εij ,

where εi ∼ N(0, V i) and V i = dJ4 + σ2I4 + τ 2Hi, with

Hi =

⎛⎜⎜⎜⎜⎝1 ρ ρ2 ρ3

ρ 1 ρ ρ2

ρ2 ρ 1 ρ

ρ3 ρ2 ρ 1

⎞⎟⎟⎟⎟⎠ .

The intercept for the placebo group is β0 + β1, and the intercept for the treatment group isβ0. The slopes are β2 and β3, respectively.

Page 357: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

344 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 12.5 Linear mixed model in the exercise study example

proc mixed data=exercise method=ml;class group id;model y=group time*group/s;repeated/type=ar(1) local subject=id;random intercept/type=un subject=id;run;

The parameter estimates from the fitted linear mixed model (Program 12.5) are shownin Table 12.5 and will later be used as initial values in Program 12.7.

Table 12.5 Parameter Estimates of the Linear MixedModel Used as Initial Values in Program 12.7

Parameter Estimate Initial value

β0 −0.8233 −0.82β1 0.1605 0.16β2 0.9227 0.92β3 1.6451 1.65d 2.0811 2.08τ 2 0.7912 0.79ρ 0.4639 0.46σ2 0.2311 0.23

Further, initial values for the parameters of the dropout model are also needed. Asmentioned before, we will fit three models in turn, under the MCAR (ψ1 = ψ2 = 0), MAR(ψ2 = 0) and MNAR mechanisms, respectively. The initial values for these parameters aregiven in Table 12.6.

Table 12.6 Initial Values for the Parameters of the Dropout Model

Dropout Mechanism

Parameter MCAR MAR MNAR MNAR + Covariate

ψ0 1 ψ0,MCAR ψ0,MAR ψ0,MNAR

ψ1 1 ψ1,MAR ψ1,MNAR

ψ2 1 ψ2,MNAR

γ 1

The parameters that will later be passed to Program 12.7 (X and Z matrices, Y andINITIAL vectors, and NSUB and NTIME parameters) are created using PROC IML.Program 12.6 illustrates the process of creating these variables when the missingnessmechanism is MCAR.

Program 12.6 Creating matrices, vectors and numbers necessary for Program 12.7

proc iml;use exercise;read all var {id group time y} into data;id=data[,1];group1=data[,2];group0=j(nrow(data),1,1)-group1;time=data[,3];timegroup0=time#group0;timegroup1=time#group1;

Page 358: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 345

intercept=j(nrow(data),1,1);create x var {intercept group0 timegroup0 timegroup1}; append;y=data[,4];create y var {y}; append;z=j(nrow(y),1,1);create z var {z}; append;beta=-0.82//0.16//0.92//1.65;D=2.08;tau2=0.79;rho=0.46;sigma2=0.23;psi=1;initial=beta//D//tau2//rho//sigma2//psi;create initial var {initial}; append;nsub=50;create nsub var {nsub}; append;ntime=4;create ntime var {ntime}; append;quit;

Program 12.7 fits the Diggle-Kenward model under the MCAR assumption. To savespace, the complete SAS code is provided on the book’s companion Web site.

Program 12.7 Diggle-Kenward model under the MCAR assumption in the exercise study example

proc iml;use x; read all into x;use y; read all into y;use z; read all into z;use nsub; read all into nsub;use ntime; read all into ntime;use initial; read all into initial;g=j(nsub,1,0);start integr(yd) global(psi,ecurr,vcurr,lastobs);...finish integr;

start loglik(parameters) global(lastobs,vcurr,ecurr,x,z,y,nsub,ntime,nrun,psi);...finish loglik;opt=j(1,11,0);opt[1]=1;opt[2]=5;con={. . . . 0 0 -1 0 .,

. . . . . . 1 . .};call nlpnrr(rc,est,"loglik",initial,opt,con);call nlpfdd(maxlik,grad,hessian,"loglik",est);inf=-hessian;covar=inv(inf);var=vecdiag(covar);stde=sqrt(var);create result var {est stde}; append;quit;

To fit the model under the mechanisms MAR and MNAR, we need to change only a fewlines in Program 12.7. For the model under MAR, we replace

Page 359: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

346 Pharmaceutical Statistics Using SAS: A Practical Guide

psi[1]=parameters[9];

with

psi[1:2]=parameters[9:10];

while, under the MNAR assumption, it is replaced by

psi[1:3]=parameters[9:11];

Further, under the MAR and MNAR assumptions, we add one or two columns of dots,respectively, to the constraints matrix (CON). Parameter estimates resulting from themodel fitted under the three missingness mechanisms, together with the estimates of theignorable analysis using PROC MIXED, are listed in Table 12.7.

Table 12.7 Results under Ignorable, MCAR, MAR, and MNAR Assumptions with and without Covariate inthe Dropout Model (Exercise Study Example)

Dropout Mechanism

Ignorable MCAR MAR MNAR MNAR + Cov.

Par. Est. (s.e.) Est. (s.e.) Est. (s.e.) Est. (s.e.) Est. (s.e.)

β0 −0.82 (0.39) −0.82 (0.40) −0.82 (0.40) −0.83 (0.40) −0.82 (0.40)β1 0.16 (0.56) 0.16 (0.56) 0.16 (0.56) 0.17 (0.56) 0.16 (0.56)β2 0.92 (0.10) 0.92 (0.10) 0.92 (0.10) 0.93 (0.10) 0.92 (0.10)β3 1.65 (0.10) 1.65 (0.10) 1.65 (0.10) 1.66 (0.11) 1.64 (0.11)

d 2.08 (0.90) 2.08 (0.91) 2.08 (0.90) 2.09 (0.85) 2.07 (0.96)τ 2 0.79 (0.54) 0.79 (0.55) 0.79 (0.55) 0.80 (0.70) 0.79 (0.45)ρ 0.46 (1.10) 0.46 (1.13) 0.46 (1.12) 0.44 (1.12) 0.49 (1.13)σ2 0.23 (1.08) 0.23 (1.11) 0.23 (1.11) 0.21 (1.24) 0.25 (1.02)

ψ0 −2.33 (0.30) −2.17 (0.36) −2.42 (0.88) −1.60 (1.14)ψ1 −0.10 (0.14) −0.24 (0.43) −0.018 (0.47)ψ2 0.16 (0.47) −0.13 (0.54)γ −0.66 (0.79)

−2� 641.77 641.23 641.11 640.44

Analysis of Exercise Study Data (Extended Model)Next, we extend the Diggle-Kenward model by allowing the dropout process to depend ona covariate (GROUP). Thus, instead of (12.7.16), we use the following model

logit [P (Di = j | Di ≥ j, hij , yij , ψ)] = ψ0 + ψ1yi,j−1 + ψ2yij + γ groupi. (12.7.18)

Program 12.7 can easily be adapted to this case. First, in the INTEGR and LOGLIKmodules, we add GROUP and GROUPI as global variables. Further, in the LOGLIKmodule, the γ parameter is specified as well:

group=parameters[12];

and where the information on a particular patient is selected, we add:

groupi = xi[1,2];

Next, in the INTEGR module, we replace

Page 360: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 347

g=exp(psi[1]+psi[2]*lastobs+psi[3]*yd);

with

g=exp(psi[1]+psi[2]*lastobs+psi[3]*yd+group*groupi);

and in the LOGLIK module,

g = exp(psi[1]+yobs[j-1]*psi[2]+yobs[j]*psi[3]);

is replaced by

g = exp(psi[1]+yobs[j-1]*psi[2]+yobs[j]*psi[3]+group*groupi);

Finally, as in the MNAR program, we again add a column of dots to the constraints matrix(CON). The initial values used are listed in Table 12.6. The results produced by theprogram are displayed in Table 12.7. The results of the measurement model should be thesame under the ignorable, MCAR, and MAR assumptions. As we can see from Table 12.7,this is more or less the case, except for some of the variance components, due to slightnumerical variation. Adding the covariate to the dropout model results in a deviancechange of 0.67, which means the covariate is not significant (p-value is 0.413). Alikelihood-ratio test for the MAR versus MNAR assumption (ψ2 = 0 or not) will not befully trustworthy (Jansen et al., 2005). Note that, under the MNAR assumption, theestimates for ψ1 and ψ2 are more or less equal, but with different signs.

12.8 Sensitivity AnalysisSensitivity to model assumptions has been reported for about two decades (Verbeke andMolenberghs, 2000; Molenberghs and Verbeke, 2005). In an attempt to formulate an answerto these concerns, a number of authors have proposed strategies to study sensitivity.

Broadly, we could define a sensitivity analysis as one in which several statistical modelsare considered simultaneously and/or where a statistical model is further scrutinized usingspecialized tools (such as diagnostic measures). This rather loose and very generaldefinition encompasses a wide variety of useful approaches. The simplest procedure is to fiteither a selected number of (MNAR) models which are all deemed plausible or to fit onemodel in which a preferred (primary) analysis is supplemented with a number of variations.The extent to which conclusions (inferences) are stable across such ranges provides anindication about the belief that can be put into them. Variations to a basic model can beconstructed in different ways. The most obvious strategy is to consider variousdependencies of the missing data process on the outcomes and/or on covariates.Alternatively, the distributional assumptions of the models can be changed.

Several publications have proposed the use of global and local influence tools (Verbekeet al., 2001; Verbeke and Molenberghs, 2000; Molenberghs and Verbeke, 2005). Animportant question is this: To what exactly are the sources causing an MNAR model toprovide evidence for MNAR against MAR? There is evidence enough for us to believe thata multitude of outlying aspects is responsible for an apparent MNAR mechanism (Jansenet al., 2005). But it is not necessarily believable that the (outlying) nature of themissingness mechanism in one or a few subjects is responsible for an apparent MNARmechanism. The consequence of this is that local influence should be applied andinterpreted with due caution.

Further, within the selection model framework, Baker, Rosenberger, and DerSimonian(1992) proposed a model for multivariate and longitudinal binary data, subject tononmonotone missingness. Jansen et al. (2003) extended this model to allow for (possiblycontinuous) covariates, and developed a local influence strategy.

Page 361: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

348 Pharmaceutical Statistics Using SAS: A Practical Guide

Next, classical inference procedures account for the imprecision resulting from thestochastic component of the model. Less attention is devoted to the uncertainty arisingfrom (unplanned) incompleteness in the data, even though the majority of clinical studiessuffer from incomplete follow-up. Molenberghs et al. (2001) acknowledge both the status ofimprecision, due to (finite) random sampling, as well as ignorance, due to incompleteness.Further, both can be combined into uncertainty (Kenward, Molenberghs, and Goetghebeur,2001).

Another route for sensitivity analysis is to consider pattern-mixture models as acomplement to selection models. A third framework consists of so-called shared parametermodels, where random effects are employed to describe the relationship between themeasurement and dropout processes (Wu and Carroll, 1988; DeGruttola and Tu, 1994).

More detail on some of these procedures can be found in Molenberghs and Verbeke(2005). Let us now turn to the case of sensitivity analysis tools for selection models.

Sensitivity Analysis for Selection ModelsParticularly within the selection modeling framework, there has been an increasingliterature on MNAR missingness. At the same time, concern has been growing preciselyabout the fact that models often rest on strong assumptions and relatively little evidencefrom the data themselves.

A sensible compromise between blindly shifting to MNAR models or ignoring themaltogether is to make them a component of a sensitivity analysis. In any case, fitting anMNAR dropout model should be subject to careful scrutiny. The modeler needs to payattention, not only to the assumed distributional form of the model (Little, 1994b;Kenward, 1998), but also to the impact one or a few influential subjects may have on thedropout and/or measurement model parameters. Because fitting an MNAR dropout modelis feasible by virtue of strong assumptions, such models are likely to pick up a wide varietyof influences in the parameters describing the nonrandom part of the dropout mechanism.Hence, a good level of caution is in place.

First, an informal sensitivity analysis is applied on the mastitis data. Next, the model ofDiggle and Kenward (1994) is adapted to a form useful for sensitivity analysis, whereaftersuch a sensitivity analysis method, based on local influence (Cook, 1986; Thijs,Molenberghs and Verbeke, 2000), is introduced and applied to the mastitis data.

Informal Sensitivity Analysis of the Mastitis Data

Diggle and Kenward (1994) and Kenward (1998) performed several analyses of the mastitisdata described in Section 12.2. In Diggle and Kenward (1994), a separate mean for eachgroup defined by the year of first lactation and a common time effect was considered,together with an unstructured 2 × 2 covariance matrix. The dropout model included bothYi1 and Yi2 and was reparameterized in terms of the size variable (Yi1 + Yi2)/2 and theincrement Yi2 − Yi1. Kenward (1998) carried out what we could term a data-drivensensitivity analysis. The right panel of Figure 12.3 reveals that there appear to be twocows, #4 and #5 (black dots), with unusually large increments. Kenward conjectured thatthis might mean that these animals were ill during the first lactation year, producing anunusually low yield, whereas a normal yield was obtained during the second year. A simplemultivariate Gaussian linear model is used to represent the marginal milk yield in the twoyears (i.e., the yield that would be, or was, observed in the absence of mastitis):(

Yi1

Yi2

)= N

((β0

β1

),

(σ2

1 σ12

σ12 σ22

)). (12.8.19)

Page 362: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 349

Note that β1 represents the change in average yield between the two years. Theprobability of mastitis is assumed to follow the logistic regression model:

logit [P (dropout)] = ψ0 + ψ1y1 + ψ2y2. (12.8.20)

The combined response/dropout model was fitted to the milk yields by using a programanalogous to Program 12.7, presented in Section 12.7. In addition, the MAR model(ψ2 = 0) was fitted in the same way. These fits produced the parameter estimates displayedin the All column of Table 12.8.

Using the likelihoods to compare the fit of the two models, we get a differenceG2 = 3.12. The corresponding tail probability from the χ2

1 is 0.07. This test essentiallyexamines the contribution of ψ2 to the fit of the model. Using the Wald statistic for thesame purpose gives a statistic of (−2.52)2/0.86 = 7.38, with corresponding χ2

1 probability of0.007. The discrepancy between the results of the two tests suggests that the asymptoticapproximations on which these tests are based are not very accurate in this setting, andthe standard error probability underestimates the true variability of the estimate of ψ2.

Table 12.8 Maximum Likelihood Estimates (Standard Errors) of MAR and MNARDropout Models under Several Deletion Schemes in the Mastitis Example

MAR Dropout

Parameter All (7,53,54,66,69,70) (4,5)

β0 5.77 (0.09) 5.65 (0.09) 5.81 (0.09)β1 0.71 (0.11) 0.67 (0.10) 0.64 (0.09)

σ21 0.87 (0.12) 0.72 (0.10) 0.77 (0.11)

σ12 0.63 (0.13) 0.44 (0.10) 0.72 (0.13)σ2

2 1.31 (0.20) 1.00 (0.16) 1.29 (0.20)

ψ0 −3.33 (1.52) −4.03 (1.76) −3.09 (1.57)ψ1 0.38 (0.25) 0.50 (0.30) 0.34 (0.26)ψ2 0 0 0

−2� 624.13 552.24 574.19

MNAR Dropout

Parameter All (7,53,54,66,69,70) (4,5)

β0 5.77 (0.09) 5.65 (0.09) 5.81 (0.09)β1 0.32 (0.14) 0.36 (0.15) 0.64 (0.14)

σ21 0.87 (0.12) 0.72 (0.10) 0.77 (0.11)

σ12 0.55 (0.13) 0.38 (0.11) 0.72 (0.13)σ2

2 1.57 (0.28) 1.16 (0.23) 1.29 (0.20)

ψ0 −0.34 (2.33) −0.55 (2.57) −3.10 (1.74)ψ1 2.36 (0.79) 2.01 (0.82) 0.32 (0.72)ψ2 −2.52 (0.86) −2.09 (0.98) 0.01 (0.72)

−2� 624.13 551.54 574.19

G2 for MNAR 3.12 0.70 0.0004

The dropout model estimated from the MNAR setting is as follows:

logit [P (mastitis)] = −0.34 + 2.36y1 − 2.52y2.

Some insight into this fitted model can be obtained by rewriting it in terms of the milkyield totals (Y1 + Y2) and increments (Y2 − Y1):

logit [P (mastitis)] = −0.34 − 0.078(y1 + y2) − 2.438(y2 − y1).

Page 363: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

350 Pharmaceutical Statistics Using SAS: A Practical Guide

The probability of mastitis increases with larger negative increments, i.e., animals thatshowed (or would have shown) a greater decrease in yield over the two years have a higherprobability of getting mastitis. The other differences in parameter estimates between thetwo models are consistent with this: the MNAR dropout model predicts a smaller averageincrement in yield (β1), with larger second year variance and smaller correlation caused bygreater negative imputed differences between yields.

12.8.1 Local InfluenceThe local influence approach, suggested by Cook (1986), can be used to investigate theeffect of extending an MAR model for dropout in the direction of MNAR dropout (Verbekeet al., 2001).

Again we consider the Diggle and Kenward (1994) model described in Section 12.7.1 forcontinuous longitudinal data subject to dropout. Since no data would be observedotherwise, we assume that the first measurement Yi1 is obtained for every subject in thestudy. We denote the probability of dropout at occasion k, given the subject was still in thestudy up to occasion k by g(hik, yik). For the dropout process, we now consider anextension of model (12.7.16), which can be written as

logit [g(hik, yik)] = logit [P(Di = k|Di ≥ k, yi)] = hikψ + ωyij , (12.8.21)

in which hik is the vector containing the history H ik as well as covariates. When ω equalszero and the model assumptions made are correct, the dropout model is MAR, and allparameters can be estimated using standard software since the measurement and dropoutmodel can then be fitted separately. If ω = 0, the dropout process is assumed to be MNAR.Now, a dropout model may be found to be MNAR solely because one or a few influentialsubjects have driven the analysis. To investigate sensitivity of estimation of quantities ofinterest, such as treatment effect, growth parameters, or the dropout model parameters,with respect to assumptions about the dropout model, we consider the following perturbedversion of (12.8.21):

logit [g(hik, yik)] = logit [P(Di = k|Di ≥ k, yi, Wi)] = hikψ + ωiyik, i = 1, . . . , N. (12.8.22)

There is a fundamental difference with model (12.8.21) since the ωi should not be viewed asparameters: they are local, individual-specific perturbations around a null model. In ourcase, the null model will be the MAR model, corresponding to setting ω = 0 in (12.8.21).Thus the ωi are perturbations that will be used only to derive influence measures (Cook,1986).

This scheme enables studying the effect of how small perturbation in the MNARdirection can have a large impact on key features of the model. Practically, one way ofdoing this is to construct local influence measures (Cook, 1986). Clearly, not all possibleforms of impact resulting from sensitivity to dropout model assumptions will be found inthis way, and the method proposed here should be viewed as one component of asensitivity analysis (e.g. Molenberghs, Kenward, and Goetghebeur, 2001).

When small perturbations in a specific ωi lead to relatively large differences in themodel parameters, it suggests that the subject is likely to drive the conclusions.

Cook (1986) suggests that more confidence can be put in a model which is relativelystable under small modifications. The best known perturbation schemes are based on casedeletion (Cook and Weisberg 1982) in which the study of interest is the effect of completelyremoving cases from the analysis. A quite different paradigm is the local influence approachwhere the investigation concentrates on how the results of an analysis are changed undersmall perturbations of the model. In the framework of the linear mixed model Beckman,Nachtsheim, and Cook (1987) used local influence to assess the effect of perturbing theerror variances, the random-effects variances, and the response vector. In the same context,

Page 364: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 351

Lesaffre and Verbeke (1998) have shown that the local influence approach is also useful forthe detection of influential subjects in a longitudinal data analysis. Moreover, since theresulting influence diagnostics can be expressed analytically, they often can be decomposedin interpretable components, which yield additional insights in the reasons why somesubjects are more influential than others.

We are interested in the influence of MNAR dropout on the parameters of interest. Thiscan be done in a meaningful way by considering (12.8.22) as the dropout model. Indeed,ωi = 0 for all i corresponds to an MAR process, which cannot influence the measurementmodel parameters. When small perturbations in a specific ωi lead to relatively largedifferences in the model parameters, then this suggests that these subjects may have alarge impact on the final analysis. However, even though we may be tempted to concludethat such subjects drop out non-randomly, this conclusion is misguided since we are notaiming to detect (groups of) subjects that drop out non-randomly but rather subjects thathave a considerable impact on the dropout and measurement model parameters. Indeed, akey observation is that a subject that drives the conclusions towards MNAR may be doingso, not only because its true data-generating mechanism is of an MNAR type, but also fora wide variety of other reasons, such as an unusual mean profile or autocorrelationstructure. Earlier analyses have shown that this may indeed be the case. Likewise, it ispossible that subjects, deviating from the bulk of the data because they are generatedunder MNAR, go undetected by this technique. This reinforces the concept that we mustreflect carefully upon which anomalous features are typically detected and which onestypically go unnoticed.

Key ConceptsLet us now introduce the key concepts of local influence. We denote the log-likelihoodfunction corresponding to model (12.8.22) by

�(γ|ω) =N∑

i=1

�i(γ|ωi),

in which �i(γ|ωi) is the contribution of the ith individual to the log-likelihood, and whereγ = (θ, ψ) is the s-dimensional vector, grouping the parameters of the measurement modeland the dropout model, not including the N × 1 vector ω = (ω1, ω2, . . . , ωN)′ of weightsdefining the perturbation of the MAR model. It is assumed that ω belongs to an opensubset Ω of IRN . For ω equal to ω0 = (0, 0, . . . , 0)′, �(γ|ω0) is the log-likelihood functionwhich corresponds to a MAR dropout model.

Let γ be the maximum likelihood estimator for γ, obtained by maximizing �(γ|ω0), andlet γω denote the maximum likelihood estimator for γ under �(γ|ω). The local influenceapproach now compares γω with γ. Similar estimates indicate that the parameter estimatesare robust with respect to perturbations of the MAR model in the direction of non-randomdropout. Strongly different estimates suggest that the estimation procedure is highlysensitive to such perturbations, which suggests that the choice between an MAR model anda non-random dropout model highly affects the results of the analysis. Cook (1986)proposed to measure the distance between γω and γ by the so-called likelihooddisplacement, defined by

LD(ω) = 2[�(γ|ω0) − �(γω|ω0)].

This takes into account the variability of γ. Indeed, LD(ω) will be large if �(γ|ω0) isstrongly curved at γ, which means that γ is estimated with high precision, and smallotherwise. Therefore, a graph of LD(ω) versus ω contains essential information on theinfluence of perturbations. It is useful to view this graph as the geometric surface formedby the values of the N + 1 dimensional vector ξ(ω) = (ω′, LD(ω))′ as ω varies throughout Ω.

Page 365: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

352 Pharmaceutical Statistics Using SAS: A Practical Guide

Since this influence graph can be depicted only when N = 2, Cook (1986) proposed tolook at local influence, i.e., at the normal curvatures Ch of ξ(ω) in ω0, in the direction ofsome N dimensional vector h of unit length. Let Δi be the s dimensional vector defined by

Δi =∂2�i(γ|ωi)

∂ωi∂γ

∣∣∣∣γ=γ,ωi=0

and define Δ as the (s × N) matrix with Δi as its ith column. Further, let L denote the(s × s) matrix of second order derivatives of �(γ|ω0) with respect to γ, also evaluated atγ = γ. Cook (1986) has then shown that Ch can be easily calculated by

Ch = 2|h′Δ′L−1Δh|.

Obviously, Ch can be calculated for any direction h. One evident choice is the vector hi

containing one in the ith position and zero elsewhere, corresponding to the perturbation ofthe ith weight only. This reflects the influence of allowing the ith subject to drop outnon-randomly, while the others can drop out only at random. The corresponding localinfluence measure, denoted by Ci, then becomes Ci = 2|Δ′

iL−1Δi|. Another important

direction is the direction hmax of maximal normal curvature Cmax. It shows how to perturbthe MAR model to obtain the largest local changes in the likelihood displacement. It isreadily seen that Cmax is the largest eigenvalue of −2 Δ′L−1Δ, and that hmax is thecorresponding eigenvector.

Local Influence in the Diggle-Kenward Model

As discussed in the previous section, calculation of local influence measures merely reducesto evaluation of Δ and L. Expressions for the elements of L in case Σi = σ2I are given byLesaffre and Verbeke (1998), and can easily be extended to the more general caseconsidered here. Further, it can be shown that the components of the columns Δi of Δ aregiven by

∂2�iω

∂θ∂ωi

∣∣∣∣ωi=0

= 0,∂2�iω

∂ψ∂ωi

∣∣∣∣ωi=0

= −ni∑

j=2

hijyijg(hij)[1 − g(hij)], (12.8.23)

for complete sequences (no dropout) and by

∂2�iω

∂θ∂ωi

∣∣∣∣ωi=0

= [1 − g(hid)]∂λ(yid|hid)

∂θ, (12.8.24)

∂2�iω

∂ψ∂ωi

∣∣∣∣ωi=0

= −d−1∑j=2

hijyijg(hij)[1 − g(hij)]

−hidλ(yid|hid)g(hid)[1 − g(hid)]. (12.8.25)

for incomplete sequences. All the above expressions are evaluated at γ, and whereg(hij) = g(hij , yij)|ωi=0, is the MAR version of the dropout model.

Let Vi,11 be the predicted covariance matrix for the observed vector (yi1, . . . , yi,d−1)′, Vi,22is the predicted variance for the missing observation yid, and Vi,12 is the vector of predictedcovariances between the elements of the observed vector and the missing observation. Itthen follows from the linear mixed model (12.7.15) that the conditional expectation for theobservation at dropout, given the history, equals

λ(yid|hid) = λ(yid) + Vi,12V−1i,11[hid − λ(hid)], (12.8.26)

which is used in (12.8.24).

Page 366: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 353

The derivatives of (12.8.26) with respect to the measurement model parameters are

∂λ(yid|hid)∂β

= xid − Vi,12V−1i,11Xi,(d−1), (12.8.27)

∂λ(yid|hid)∂α

=[∂Vi,12

∂α− Vi,12V

−1i,11

∂Vi,11

∂α

]V −1

i,11[hid − λ(hid)] (12.8.28)

where x′id is the dth row of Xi, and where Xi,(d−1) indicates the first (d − 1) rows Xi.

Further, α indicates the subvector of covariance parameters within the vector θ.In practice, the parameter θ in the measurement model is often of primary interest.

Since L is block-diagonal with blocks L(θ) and L(ψ), we can write for any unit vector h,Ch = Ch(θ) + Ch(ψ). It now immediately follows from (12.8.24) that influence on θ onlyarises from those measurement occasions at which dropout occurs. In particular, fromexpression (12.8.24), it is clear that the corresponding contribution is large only if (1) thedropout probability is small but the subject disappears nevertheless and (2) the conditionalmean strongly depends on the parameter of interest. This implies that complete sequencescannot be influential in the strict sense (Ci(θ) = 0) and that incomplete sequences onlycontribute only at the actual dropout time.

Implementation of Local Influence Analysis in SASProgram 12.8 relies on SAS/IML to calculate the normal curvature Ch of ξ(ω) in ω0, in thedirection the unit vector h.

As in Program 12.7, we first need to create the X and Z matrices, Y and INITIALvectors, and NSUB and NTIME parameters (this can be accomplished in PROC IML usinga program analogous to Program 12.6). The initial parameters should be the estimates ofthe parameters of the MAR model, fitted using a program analogous to Program 12.7.Next, we need the INTEGR and LOGLIK modules introduced in Program 12.7. These arecalled to calculate the log-likelihood function of the Diggle and Kenward (1994) modelunder the MNAR assumption. This is needed for the evaluation of Δ as well as for L.Program 12.8 also calls the DELTA module to calculate the Δ vector, whereas L iscalculated using the NLPFDD module of SAS/IML which was introduced earlier inSection 12.7.2. Finally, Program 12.8 created the C MATRIX data set that contains thefollowing normal curvatures in the direction of the unit vector hi containing one in the ithposition and zero elsewhere,

C = Ci, C1 = Ci(β), C2 = Ci(α), C12 = Ci(θ), C3 = Ch(ψ),

and the normal curvature in the direction of HMAX= hmax of maximal normal curvatureCMAX= Cmax. The C MATRIX data set can now be used to picture the local influencemeasures. The complete SAS code of Program 12.8 is provided on the book’s companionWeb site.

Program 12.8 Local influence sensitivity analysis

proc iml;use x; read all into x;use y; read all into y;use nsub; read all into nsub;use ntime; read all into ntime;use initial; read all into initial;g=j(nsub,1,0);start integr(yd) global(psi,ecurr,vcurr,lastobs);...finish integr;

Page 367: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

354 Pharmaceutical Statistics Using SAS: A Practical Guide

start loglik(parameters) global(lastobs,vcurr,ecurr,x,z,y,nsub,ntime,nrun,psi);...finish loglik;

start delta(parameters) global(lastobs,vcurr,ecurr,x,z,y,nsub,ntime,nrun,psi);...finish delta;

opt=j(1,11,0);opt[1]=1;opt[2]=5;con={. . 0 . 0 . . ,

. . . . . . . };call nlpnrr(rc,est,"loglik",initial,opt,con);

* Calculation of the Hessian;...* Calculation of the C-matrix;...create c_matrix var {subject ci c12i c3i c1i c2i hmax cmax};append;quit;

Mastitis DataWe will apply the local influence method to the mastitis data described in Section 12.2. Forthe measurement process, we use model (12.8.19) and thus the covariance matrix V i isunstructured. Since there are only two measurement occasions, Yi1 and Yi2, the componentsof the columns Δi of Δ are given by (12.8.23), when both measurements are available, andby (12.8.24) and (12.8.25), when only the first measurement is taken. In the latter case, weneed Vi,11, Vi,12, and their derivatives with respect to the three variance components σ2

1 , σ12and σ2

2 , to calculate (12.8.26), (12.8.27), and (12.8.28). Since an incomplete sequence canoccur only when the cow dropped out at the second measurement occasion, we have thatVi,11 = σ2

1 and Vi,12 = σ12, and thus

∂Vi,11

∂σ12=

∂Vi,12

∂σ22

= 0,∂Vi,12

∂σ21

=∂Vi,12

∂σ22

= 0,∂Vi,11

∂σ21

=∂Vi,12

∂σ12= 1.

If we have another form of model (12.7.15), the program can be adapted, by changing V i,Vi,11, Vi,12, and the derivatives.

The results of the local influence analysis of the mastitis data (based on Program 12.8)are shown in Figure 12.7 which suggests that there are six influential subjects: #7, #53,#54, #66, #69, and #70, while #4 and #5 are not recovered. It is interesting to consideran analysis with these six cows removed. The influence on the likelihood ratio test is a littlesmaller then in the case of removing #4 and #5: G2 = 0.07 compared to G2 = 0.0004 whenremoving #4 and #5 instead of the original 3.12. The influence on the measurement modelparameters under both random and non-random dropout is small for the fixed effectsparameters, but not so small for the covariance components parameters.

Local and Global InfluenceLet us now discuss differences between local and global influence. It is very important torealize that we should not expect agreement between the deletion and local influenceanalyses. The latter focuses on the sensitivity of the results with respect to the assumeddropout model, more specifically on how the results change when the MAR model is

Page 368: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 355

Figure 12.7 Index plots of Ci, Ci(θ), Ci(α), Ci(β), Ci(ψ) and of the components of the direction hmax,i ofmaximal curvature in the mastitis example

0 20 40 60 80 100

0.0

0.5

1.0

1.5

2.0

# 7

# 53

# 54

# 66

# 69

# 70

Ci

0 20 40 60 80 100

0.0

0.01

0.02

0.03

Ci (θ)

0 20 40 60 80 100

0.0

0.00

50.

010

0.01

50.

020

0.02

5

Ci (α)

0 20 40 60 80 100

0.0

0.00

20.

004

0.00

60.

008

0.01

00.

012

Ci (β)

0 20 40 60 80 100

0.0

0.5

1.0

1.5

2.0

# 7

# 53

# 54

# 66

# 69

# 70

Ci (ψ)

0 20 40 60 80 100

-1.0

-0.5

0.0

0.5

1.0

# 7

# 53

# 54

# 66

# 69

# 70

hmax,i

extended into the direction of non-random dropout. In Ci(β) and Ci(α) panels, little or noinfluence is detected, certainly when the scale is compared to the one of Ci(ψ). This isobvious for subjects #53, #54, #66, and #69, since all these are complete and henceCi(θ) ≡ 0, placing all influence on Ci(ψ). Of course, a legitimate concern is precisely wherewe should place a cut-off between subjects that are influential and those that are not.Clearly, additional work studying the stochastic behavior of the influence measures wouldbe helpful. Meanwhile, informal guidelines can be used, such as studying 5% of therelatively most influential subjects.

Kenward (1998) observed that Cows #4 and #5 in the mastitis example are unusual onthe basis of their increment. This is in line with several other applications of similardropout models (Molenberghs, Kenward, and Lesaffre, 1997) where it was found that astrong incremental component apparently yields a non-random dropout model. From the

Page 369: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

356 Pharmaceutical Statistics Using SAS: A Practical Guide

analysis done, it is clear that such a conclusion may not be well founded, since removal of#4 and #5 leads to disappearance of the non-random component (see Table 12.8).

12.9 SummaryAnalyzing incomplete (longitudinal) data, both of a Gaussian as well as of a non-Gaussiannature, can easily be conducted under the relatively relaxed assumption of missingness atrandom (MAR), using standard statistical software tools. Likelihood-based methodsinclude the linear mixed model and generalized linear mixed models. In addition, weightedgeneralized estimating equations (WGEE) can be used as a relatively straightforward fixup of ordinary generalized estimating equations (GEE) so that also this technique is validunder MAR. Alternative methods which allow for ignoring the missing data mechanismunder MAR include multiple imputation (MI) and the Expectation-Maximization (EM)algorithm. The GLIMMIX macro and PROC GLIMMIX were both presented as two usefulimplementations of the generalized estimating equations approach, usefully supplementingthe more familiar GENMOD procedure. Further, the GLIMMIX tools are useful forgeneralized linear mixed modeling, supplementing PROC NLMIXED. The depression trialwas used as an illustration.

These considerations imply that traditionally popular but very restricted modes ofanalysis, including complete case (CC) analysis, last observation carried forward (LOCF),or other simple imputation methods, ought to be abandoned, given the highly restrictiveassumptions on which they are based.

Of course, general missingness not at random can never be entirely excluded, and oneshould therefore ideally supplement an ignorable analysis with a suitable chosen set ofsensitivity analyses. Therefore, we presented a formal selection modeling framework forcontinuous outcomes, for categorical responses, and contrasted these to thepattern-mixture framework. The general MNAR selection model of Diggle and Kenward(1994) for the exercise bike data was fitted using SAS/IML code. Further, we showedexplicit sensitivity analysis methods. We presented a local influence analysis for themastitis data, supplemented with the necessary SAS/IML code. We further sketchedframeworks for incomplete binary and ordinal data, based on local influence ideas. Finally,as a sensitivity analysis for pattern-mixture models, identifying restrictions are proposed asa viable strategy.

ReferencesAkaike, H. (1974). “A new look at the statistical model identification.” IEEE Transaction on

Automatic Control. 19, 716–723.Baker, S.G., Rosenberger, W.F., DerSimonian, R. (1992). “Closed-form estimates for missing counts

in two-way contingency tables.” Statistics in Medicine. 11, 643–657.Beckman, R.J., Nachtsheim, C.J., Cook, R.D. (1987). “Diagnostics for mixed-model analysis of

variance.” Technometrics. 29, 413–426.Beunckens, C., Molenberghs, G., Kenward, M.G. (2005). “Direct likelihood analysis versus simple

forms of impuation for missing data in randomized clinical trials.” Clinical Trials. 2, 379–386.Breslow, N.E., Clayton, D.G. (1993). “Approximate inference in generalized linear mixed models.”

Journal of American Statistical Association. 88, 9–25.Burnham, K. P., Anderson, D.R. (1998). Model selection and inference, a pratical

information-theoretic approach. New York: Springer.Cook, R.D. (1986). “Assessment of local influence.” Journal of the Royal Statistical Society, Series

B. 48, 133–169.Cook, R.D., Weisberg, S. (1982). Residuals and Influence in Regression. London: Chapman and Hall.

Page 370: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 357

DeGruttola, V., Tu, X.M. (1994). “Modelling progression of CD4 lymphocyte count and itsrelationship to survival time.” Biometrics. 50, 1003–1014.

Dempster, A.P., Laird, N.M., Rubin, D. B. (1977). “Maximum likelihood from incomplete data viathe EM algorithm (with discussion).” Journal of the Royal Statistical Society, Series B. 39, 1–38.

Diggle, P.J., Kenward, M.G. (1994). “Informative drop-out in longitudinal data analysis (withdiscussion).” Applied Statistics. 43, 49–93.

Dmitrienko, A., Molenberghs, G., Chuang-Stein, C., Offen W. (2005). Analysis of Clinical TrialsUsing SAS: A Practical Guide. Cary, NC: SAS Press.

Glynn, R.J., Laird, N.M., Rubin, D.B. (1986). “Selected modelling versus mixture modelling withnon-ignorable nonresponse.” Drawing Inferences from Self Selected Samples, H. Wainer (ed.).New York: Springer-Verlag, 115–142.

Heyting, A., Tolboom, J., Essers, J. (1992). “Statistical handling of dropouts in longitudinal clinicaltrials.” Statistics in Medicine. 11, 2043–2061.

Hogan, J.W. and Laird, N.M. (1997). “Mixture models for the joint distribution of repeatedmeasures and event times.” Statistics in Medicine. 16, 239–258.

Jansen, I., Hens, N., Molenberghs, G., Aerts, M., Verbeke, G., Kenward, M.G. (2006). “The natureof sensitivity in monotone missing not at random models.” Computational Statistics and DataAnalysis. 50, 830–858.

Jansen, I., Molenberghs, G., Aerts, M., Thijs, H., Van Steen, K. (2003). “A local influence approachapplied to binary data from a psychiatric study.” Biometrics. 59, 409–418.

Kenward, M.G. (1998). “Selection models for repeated measurements with nonrandom dropout: anillustration of sensitivity.” Statistics in Medicine. 17, 2723–2732.

Kenward, M.G., Molenberghs, G., Goetghebeur, E. (2001). “Sensitivity analysis for incompletecategorical data.” Statistical Modelling. 1, 31–48.

Lavori, P.W., Dawson, R., Shera, D. (1995). “A multiple imputation strategy for clinical trials withtruncation of patient data.” Statistics in Medicine. 14, 1913–1925.

Lesaffre, E., Verbeke, G. (1998). “Local influence in linear mixed models.” Biometrics. 54, 570–582.Liang, K.Y., Zeger, S.L. (1986). “Longitudinal data analysis using generalized linear models.”

Biometrika. 73, 13–22.Little, R.J.A. (1993). “Pattern-mixture models for multivariate incomplete data.” Journal of

American Statistical Association. 88, 125–134.Little, R.J.A. (1994a). “A class of pattern-mixture models for normal incomplete data.” Biometrika.

81, 471–483.Little, R.J.A. (1994b). “Discussion to Diggle, P.J., Kenward, M.G. Informative dropout in

longitudinal data analysis.” Applied Statistics. 43, 78.Little, R.J.A., Rubin, D.B. (1987). Statistical Analysis with Missing Data. New York: Wiley.Mallinckrodt C.H., Clark W.S., Carroll R.J., Molenberghs G. (2003a). “Assessing response profiles

from incomplete longitudinal clinical trial data under regulatory considerations.” Journal ofBiopharmaceutical Statistics. 13, 179–190.

Mallinckrodt, C.H., Clark, W.S., David, S. (2001a). “Type I error rates from mixed-effects modelrepeated measures versus fixed effects analysis of variance with missing values imputed via lastobservation carried forward.” Drug Information Journal. 35, 4, 1215–1225.

Mallinckrodt, C.H., Clark, W.S., David, S. (2001b). “Accounting for dropout bias usingmixed-effects models.” Journal of Biopharmaceutical Statistics. 11, 9–21.

Mallinckrodt, C.H., Sanger, T.M., Dube, S., Debrota, D.J., Molenberghs, G., Carroll, R.J., ZeiglerPotter, W.M., Tollefson, G.D. (2003b). “Assessing and interpreting treatment effects inlongitudinal clinical trials with missing data.” Biological Psychiatry. 53, 754–760.

McCullagh, P., Nelder, J.A. (1989). Generalized Linear Models. London: Chapman and Hall.McLachlan, G.J., Krishnan, T. (1997). The EM algorithm and extensions. New York: John Wiley

and Sons.Molenberghs, G., Goetghebeur, E., Lipsitz S.R., Kenward, M.G. (1999). “Non-random missingness

in categorical data: strengths and limitations.” The American Statistician. 53, 110–118.

Page 371: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

358 Pharmaceutical Statistics Using SAS: A Practical Guide

Molenberghs, G., Kenward, M.G., Goetghebeur, E. (2001). “Sensitivity analysis for incompletecontingency tables: the Slovenian plebiscite case.” Journal of the Royal Statistical Society, SeriesC: Applied Statistics. 50, 15–29.

Molenberghs, G., Kenward, M.G., Lesaffre, E. (1997). “The analysis of longitudinal ordinal datawith nonrandom dropout.” Biometrika. 84, 33–44.

Molenberghs, G., Thijs, H., Jansen, I., Beunckens, C., Kenward, M.G., Mallinckrodt, C., Carroll,R.J. (2004). “Analyzing incomplete longitudinal clinical trial data.” Biostatistics. 5, 445–464.

Molenberghs, G., Verbeke, G. (2004). “Meaningful statistical model formulations for repeatedmeasures.” Statistica Sinica. 14, 989–1020.

Molenberghs, G., Verbeke, G. (2005). Models for Discrete Longitudinal Data. New York: Springer.Molenberghs, G., Verbeke, G., Thijs, H., Lesaffre, E., Kenward, M.G. (2001). “Mastitis in dairy

cattle: influence analysis to assess sensitivity of the dropout process.” Computational Statisticsand Data Analysis. 37, 93–113.

Murray G.D., Findlay J.G. (1988). “Correcting for the bias caused by drop-outs in hypertensiontrials.” Statististics in Medicine. 7, 941–946.

Nelder, J.A., Mead, R. (1965). “A simplex method for function minimisation.” The ComputerJournal. 7, 303–313.

Nelder, J.A., Wedderburn, R.W.M. (1972). “Generalized linear models.” Journal of the RoyalStatistical Society, Series B. 135, 370–384.

Neuhaus, J.M. (1992). “Statistical methods for longitudinal and clustered designs with binaryresponses.” Statistical Methods in Medical Research. 1, 249–273.

Neuhaus, J.M., Kalbfleisch, J.D., Hauck, W.W. (1991). “A comparison of cluster-specific andpopulation-averaged approaches for analyzing correlated binary data.” International StatisticalReview. 59, 25–35.

Pinheiro, J.C., Bates, D.M. (1995). “Approximations to the log-likelihood function in the nonlinearmixed-effects model.” Journal of Computational and Graphical Statistics. Series 4, 12–35.

Pinheiro, J.C., Bates, D. M. (2000). Mixed-Effects Models in S and S-PLUS, Statistics andComputing. New York: Springer.

Press, W., Teukolsky, S., Vetterling, W., Flannery, B. (1992). Numerical Recipes in C: The Art ofScientific Computing. Second edition. New York: Cambridge University Press.

Robins, J.M., Rotnitzky, A., Scharfstein, D.O. (1998). “Semiparametric regression for repeatedoutcomes with non-ignorable non-response.” Journal of American Statistical Association. 93,1321–1339.

Robins, J.M., Rotnitzky, A., Zhao, L.P. (1995). “Analysis of semiparametric regression models forrepeated outcomes in the presence of missing data.” Journal of American Statistical Association.90, 106–121.

Rubin, D.B. (1976). “Inference and missing data.” Biometrika. 63, 581–592.Rubin, D.B. (1978). “Multiple imputations in sample surveys – a phenomenological Bayesian

approach to nonresponse.” Imputation and Editing of Faulty or Missing Survey Data.Washington, DC: U.S. Department of Commerce, 1–23.

Rubin, D.B. (1987). Multiple Imputation for Nonresponse in Surveys. New York: Wiley.Rubin, D.B., Schenker, N. (1986). “Multiple imputation for interval estimation from simple random

samples with ignorable nonresponse.” Journal of the American Statistical Association. 81,366–374.

Rubin, D.B., Stern, H.S., Vehovar, V. (1995). “Handling ‘don’t know’ survey responses: the case ofthe Slovenian plebiscite.” Journal of American Statistical Association. 90, 822–828.

Schafer J.L. (1997). Analysis of Incomplete Multivariate Data. London: Chapman and Hall.Scharfstein, D.O., Rotnitzky, A., Robins, J.M. (1999). “Adjusting for nonignorable drop-out using

semiparametric nonresponse models (with discussion).” Journal of the American StatisticalAssociation. 94, 1096–1120.

Schwarz, G. (1978). “Estimating the dimension of a model.” Annals of Statistics. 6, 461–464.

Page 372: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 12 Analysis of Incomplete Data 359

Siddiqui, O., Ali, M.W. (1998). “A comparison of the random-effects pattern mixture model withlast observation carried forward (LOCF) analysis in longitudinal clinical trials with dropouts.”Journal of Biopharmaceutical Statistics. 8, 545–563.

Skellam, J.G. (1948). “A probability distribution derived from the binomial distribution byregarding the probability of success as variable between the sets of trials.” Journal of the RoyalStatistical Society, Series B. 10, 257–261.

Skrondal, A., Rabe-Hesketh, S. (2004). Generalized latent variable modeling: Multilevel, longitudinaland structural equation models. London: Chapman and Hall.

Stiratelli, R., Laird, N., Ware, J. (1984). “Random effects models for serial observations withdichotomous response.” Biometrics. 40, 961–972.

Thijs, H., Molenberghs, G., Verbeke, G. (2000). “The milk protein trial: influence analysis of thedropout process.” Biometrical Journal. 42, 617–646.

Tierney, L. Kadane, J.B. (1986). “Accurate approximations for posterior moments and marginaldensities.” Journal of the American Statistical Association. 81, 82–86.

Tuerlinckx, F., Rijmen, F., Molenberghs, G., Verbeke, G., Briggs, D., Van den Noortgate, W.,Meulders, M., De Boeck, P. (2004). “Estimation and software,” in Explanatory Item ResponseModels: A Generalized Linear and Nonlinear Approach. De Boeck, P., Wilson, M. (editors). NewYork: Springer, 343–373.

Van Steen, K., Molenberghs, G., Verbeke, G., Thijs, H. (2001). “A local influence approach tosensitivity analysis of incomplete longitudinal ordinal data.” Statistical Modelling: AnInternational Journal. 1, 125–142.

Verbeke, G., Molenberghs, G. (2000). Linear Mixed Models for Longitudinal Data. New York:Springer.

Verbeke, G., Molenberghs, G., Thijs, H., Lesaffre, E., Kenward, M.G. (2001). “Sensitivity analysisfor non-random dropout: a local influence approach.” Biometrics. 57, 43–50.

Wolfinger, R., O’Connell, M. (1993). “Generalized linear mixed models: a pseudo-likelihoodapproach.” Journal of Statistical Computation and Simulation. 48, 233–243.

Wu, M.C., Carroll, R.J. (1988). “Estimation and comparison of changes in the presence ofinformative right censoring by modeling the censoring process.” Biometrics. 44, 175–188.

Page 373: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications
Page 374: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13

Reliability and Validity:Assessing the PsychometricProperties of Rating Scales

Douglas FariesIlker Yalcin

13.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361

13.2 Reliability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .362

13.3 Validity and Other Topics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .376

13.4 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 382

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383

Using outcome measures that are reliable and valid is a prerequisite for quality clinicalresearch. In this chapter, statistical approaches for assessing the reliability and validity ofan outcome measure are presented and discussed. In addition to explanations of the generalconcepts, examples from multiple clinical experiments using a variety of outcome measuresare provided.

13.1 IntroductionAdequate clinical trials require the use of valid and reliable measurements. In general, ameasure is said to be valid if it accurately reflects the concept it is intended to measure. Ameasure is said to be reliable if repeated assessments of a stable subject or population usingthe measure produces similar results. The establishment of the reliability and validity of ameasure is not a single test, but rather it is a summary of its psychometric properties usingmultiple approaches.

Section 2.2.2 of the ICH E9 guidelines emphasizes the importance of using primarymeasures that are both reliable and valid for the population enrolled in a clinical trial.Specifically, “there should be sufficient evidence that the primary variable can provide avalid and reliable measure of some clinically relevant and important treatment benefit inthe population described by the inclusion exclusion criteria.” In addition, the guidelinesstate that when rating scales are used as primary variables, it is especially important toaddress such factors as validity, inter- and intra-rater reliability, and responsiveness.

Regulatory guidance is not the only reason to use measures with adequate reliabilityand validity. Both Kraemer (1991) and Leon et al. (1995) studied the relationship betweenreliability and power to detect treatment differences. Kraemer assessed test-retest

Douglas Faries is Research Advisor, Global Statistical Sciences, Eli Lilly and Company, USA. Ilker Yalcin is Manager,Commercialization Information Sciences, Eli Lilly and Company, USA.

Page 375: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

362 Pharmaceutical Statistics Using SAS: A Practical Guide

reliability while Leon et al. focused on internal consistency. Both demonstrated that largeincreases in power are possible (with a fixed sample size) by using a scale with greaterreliability as opposed to a measure with low to moderate reliability. Faries et al. (2001)showed that the impact of using a unidimensional rather than a multidimensional scale fordepression clinical trials results in the need for approximately one-third fewer subjects. Dueto overlapping symptoms in some disease states, using a scale with discriminant validity iscritical to making claims of efficacy for a particular disease state. In summary, use of ameasure without established reliability may prove unnecessarily costly (result in a largertrial than necessary or a failed trial due to reduced power), and using a measure withoutappropriate validity can severely affect the credibility of the results.

In drug development, one is often faced with the challenge of validating a measure orselecting a measure based in part on their psychometric properties. For instance, one maywish to use a scale that is validated as a patient-rated scale as an observer-rated scale. Inaddition, one may need to create a composite variable, use a subscale, or wish to develop anew rating scale with potential advantages over existing measures. In each of thesescenarios, the statistician is faced with the challenge of quantitatively demonstrating thereliability and validity of the outcome measures. Even when appropriate validation for ameasure has been established, often one is interested in documenting inter-rater reliabilityfor the group of individuals who will be conducting a specific experiment or trial.

In this chapter we review some basic approaches for assessing the reliability and validityof an outcome measure and provide several numerical examples using SAS. Thedevelopment of a rating scale typically starts with literature reviews, focus groups, andcognitive interviews to help develop and refine potential items. However, the developmentstage of an outcome measure is beyond the scope of this chapter. For an example of thedevelopment of a new scale, see Cappelleri et al. (2004).

To save space, some SAS code has been shortened and some output is not shown. Thecomplete SAS code and data sets used in this book are available on the book’s companionWeb site at http://support.sas.com/publishing/bbu/companion site/60622.html.

13.1.1 Other MethodsThe following reliability/validity methods are not discussed in detail in this chapter, butare provided here as a reference. For cases when a value can be specified for which adifference of less than the value is clinically non-relevant, Lin et al. (2002) discusses the useof a total deviation index and coverage probabilities relative to the concordance correlationcoefficient (CCC). Evans et al. (2004) provides an example of using item response theory toidentify items from the Hamilton Rating Scale for Depression (HAMD) with poorpsychometric properties. Beretvas and Pastor (2003) summarize the use of mixed-effectmodels and reliability generalization studies for assessing reliability. Lastly, Donner andEliasziw (1987) discuss sample size guidelines for reliability studies.

13.2 ReliabilityReliability is the degree to which multiple measurements on a subject agree. Typically thisincludes establishing appropriate levels of inter-rater reliability, test-retest reliability, andinternal consistency. Inter-rater reliability is an assessment of the agreement betweendifferent raters’ scores for the same subject at the same point in time. Test-retest reliabilitysummarizes the consistency between different measurements on the same subject over time.Intra-rater reliability is a special case of test-retest reliability where measurements areobtained by the same rater over time on the same subject. Internal consistency evaluatesthe consistency with the items of a scale assessing the same construct. In this section wediscuss several approaches to quantitatively assessing reliability, and we provide numericalexamples using SAS.

Page 376: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 363

13.2.1 Inter-Rater ReliabilityA measure is said to have inter-rater reliability if ratings by different individuals at thesame time on the same subject are similar. Inter-rater reliability is not only assessed whenvalidating a newly created instrument, but is also used prior to beginning a clinical trial toquantify the reliability of the measure as used by the raters participating in the trial. Inthe latter case, these evaluation sessions are used to both train and quantify inter-raterreliability, even when a scale with established reliability is being used.

13.2.2 Kappa StatisticThe Kappa statistic (κ) is the most commonly used tool for quantifying the agreementbetween raters when using a dichotomous or nominal scale, while the intraclass correlationcoefficient (ICC) or concordance correlation coefficient (CCC) are the recommendedstatistics for continuous measures. The use of κ will be discussed in this section whileexamples of the use of ICC and CCC are presented later (see Sections 13.2.5 and 13.2.7).

The κ statistic is the degree of agreement above and beyond chance agreementnormalized by the degree of attainable agreement above what would be predicted bychance. This measure is superior to simple percent agreement as it accounts for chanceagreement. For instance, if two raters diagnose a set of 100 patients for the presence orabsence of a rare disease, they would have a high percentage of agreement even if they eachrandomly chose one subject to classify as having the disease and classified all others as nothaving the disease. However, the high agreement would likely not result in a high valuefor κ.

Table 13.1 Notation Used in the Definitionof the κ Statistic

Rater BRater 1 Yes No Total

Yes n11 n12 n1.

No n21 n22 n2.

Total n.1 n.2 n

To define κ, consider the notation for a 2 × 2 table displayed in Table 13.1 and letpij = nij/n, pi. = ni./n and p.j = n.j/n. The value of κ can be estimated by using theobserved cell proportions as follows:

κ =pa − pc

1 − pc,

where pa is given by pa =∑

i pii and represents the observed proportion of agreement inthese data. Further, pc is defined as pc =

∑i pi.p.i and represents the proportion of

agreement expected by chance. Confidence intervals can be computed using the asymptoticstandard deviation of κ provided by Fleiss et al. (1969):

sκ =[A + B − C

(1 − pc)2n

],

where

A =∑

i

pii[1 − (pi. + p.i)(1 − κ)]2,

B = (1 − κ)2∑i �=j

pij(pi. + p.i)2,

C = [κ − pc(1 − κ)]2.

Page 377: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

364 Pharmaceutical Statistics Using SAS: A Practical Guide

Fleiss (1981) provided the variance of κ under the null hypothesis (κ = 0) which can beused to construct a large sample test statistic:

sκ0 =1√

n(1 − pc)

[pc + p2

c −∑

i

pi.p.i(pi. + p.i)

]1/2

.

For exact inference calculations, the observed results are compared with the set of allpossible tables with the same marginal row and column scores. See Agresti (1992) or Mehtaand Patel (1983) for details on computing exact tests for contingency table data.

EXAMPLE: Schizophrenia diagnosis exampleThe simplest example of inter-rater reliability study is when two raters rate a group ofsubjects as to whether certain criteria (e.g., diagnosis) are met using a diagnosticinstrument. The κ statistic is the traditional approach to assessing interrater reliability insuch circumstances. PROC FREQ provides for easy computation of the κ statistic, withinferences based on either large sample assumptions or an exact test.

Bartko (1976) provided an example where two raters classified subjects as to whetherthey had a diagnosis of schizophrenia. These data, modified for this example, are displayedin Table 13.2 and will be used to demonstrate the computation of the κ statistic in SAS.

Table 13.2 Schizophrenia DiagnosisExample Data Set (N = 30)

Rater BRater A Yes No

Yes 5 3No 1 21

Program 13.1 uses the FREQ procedure to compute the κ statistic in the schizophreniadiagnosis example.

Program 13.1 Computation of the κ statistic

data schiz;input rater_a $ rater_b $ num;cards;yes yes 5yes no 3no yes 1no no 21;

proc freq data=schiz;weight num;tables rater_a*rater_b;exact agree;run;

Output from Program 13.1

Simple Kappa Coefficient

Kappa (K) 0.6296ASE 0.166595% Lower Conf Limit 0.303395% Upper Conf Limit 0.9559

Page 378: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 365

Test of H0: Kappa = 0

ASE under H0 0.1794Z 3.5093One-sided Pr > Z 0.0002Two-sided Pr > |Z| 0.0004

Exact TestOne-sided Pr >= K 0.0021Two-sided Pr >= |K| 0.0021

Sample Size = 30

Note that in this example the observed agreement is p11 + p22 = 26/30 = 86.7%.However, a 64% agreement can be expected by chance given the observed marginals, i.e.,

1N

∑i

ni.n.i = 19.2/30 = 64%.

Output 13.1 shows that the estimated κ is 0.6296 with a 95% confidence interval of(0.3033, 0.9559). This level of agreement (in the range of 0.4 to 0.8) is considered moderate,while κ values of 0.80 and higher indicate excellent agreement (Stokes et al., 2000).Output 13.1 also provides for testing the null hypothesis of κ = 0. By including theEXACT AGREE statement in Program 13.1, we obtained both large sample and exactinferences. Both tests indicate the observed agreement is greater than chance (p = 0.0004using the z-statistic and p = 0.0021 using the exact test).

Two extensions of the κ measure of agreement, the weighted κ and stratified κ, are alsoeasily estimated using PROC FREQ. A weighted κ is applied to tables larger than 2 by 2and allows different weight values to be placed on different levels of disagreement. Forexample, on a 3-point scale, i.e., (0, 1, 2), a weighted κ would penalize cases where theraters differed by 2 points more than cases where the difference was one. Both Cicchettiand Allison (1971) and Fleiss and Cohen (1973) have published weight values for use incomputing a κ statistic. Stokes et al. (2000) provides an example of using SAS to computea weighted κ statistic.

When reliability is to be assessed across several strata, a weighted average of strataspecific κ’s can be produced. SAS uses the inverse variance weighting scheme. See thePROC FREQ documentation for details.

The κ statistic has been criticized because it is a function of not only the sensitivity(proportion of cases identified or true positive rate) and specificity (proportion of non-casescorrectly identified) of an instrument, but also of the overall prevalence of cases (base rate).For instance, Grove et al. (1981) pointed out that with sensitivity and specificity heldconstant at 0.95, a change in the prevalence can greatly influence the value of κ. Spitznageland Helzer (1985) noted that a diagnostic procedure showing a high κ in a clinical trialwith a high prevalence will have a lower value of κ in a population based study, even whenthe sensitivity and specificity are the same. Thus, comparisons of agreement across differentstudies where the base rate (prevalence) differs are problematic with the κ statistic.

13.2.3 Proportion of Positive Agreement and Proportion ofNegative Agreement

One approach to overcoming this drawback with the κ statistic is to use a pair of statistics:the proportion of positive agreement, p+, and the proportion of negative agreement, p−.Consider the situation where two raters classify a set of patients as having or not having a

Page 379: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

366 Pharmaceutical Statistics Using SAS: A Practical Guide

specific disease. The first measure, p+, is the conditional probability that a rater will give apositive diagnosis given that the other rater has given a positive diagnosis. Similarly, p− isthe conditional probability that a rater will give a negative diagnosis given that the otherrater has given a negative diagnosis. These conditional probabilities are defined as follows

p+ = 2p11/(2p11 + p12 + p21), p− = 2p22/(2p22 + p12 + p21)

and are analogous to characterizing a diagnostic test’s sensitivity (the proportion ofpatients with a disease correctly identified) and specificity (the proportion of patientswithout the disease correctly identified). Note that p+ can be viewed as a weighted averageof the sensitivities computed considering each rater as the gold standard separately, and p−can be viewed as a weighted average of the specificities computed considering each rater asthe gold standard separately. In addition, sensitivity and specificity are not even applicablein situations where there is no gold standard test. However, p+ and p− are still usefulstatistics in such situations, e.g., p+ is the conditional probability that a rater will give apositive diagnosis given that the other rater has given a positive diagnosis.

Program 13.2 demonstrates how to compute p+, p−, and associated confidence intervalsin the schizophrenia diagnosis example. The confidence intervals are computed usingformulas based on the delta method provided by Graham and Bull (1998).

Program 13.2 Computation of the proportion of positive agreement, proportion of negative agreement, andassociated confidence intervals

data agreement;set schiz nobs=m;format p_pos p_neg lower_p lower_n upper_p upper_n 5.3;if rater_a=’yes’ and rater_b=’yes’ then d=num;if rater_a=’yes’ and rater_b=’no’ then c=num;if rater_a=’no’ and rater_b=’yes’ then b=num;if rater_a=’no’ and rater_b=’no’ then a=num;n=a+b+c+d;p1=a/n; p2=b/n; p3=c/n; p4=d/n;p_pos=2*d/(2*d+b+c);p_neg=2*a/(2*a+b+c);phi1=(2/(2*p4+p2+p3))-(4*p4/((2*p4+p2+p3)**2));phi2_3=-2*p4/(((2*p4+p2+p3)**2));gam2_3=-2*p1/(((2*p1+p2+p3)**2));gam4=(2/(2*p1+p2+p3))-(4*p1/((2*p1+p2+p3)**2));sum1=(phi1*p4+phi2_3*(p2+p3))**2;sum2=(gam4*p1+gam2_3*(p2+p3))**2;var_pos=(1/n)*(p4*phi1*phi1+(p2+p3)*phi2_3*phi2_3-4*sum1);var_neg=(1/n)*(p1*gam4*gam4+(p2+p3)*gam2_3*gam2_3-4*sum2);lower_p=p_pos-1.96*sqrt(var_pos);lower_n=p_neg-1.96*sqrt(var_neg);upper_p=p_pos+1.96*sqrt(var_pos);upper_n=p_neg+1.96*sqrt(var_neg);retain a b c d;label p_pos=’Proportion of positive agreement’

p_neg=’Proportion of negative agreement’lower_p=’Lower 95% confidence limit’lower_n=’Lower 95% confidence limit’upper_p=’Upper 95% confidence limit’upper_n=’Upper 95% confidence limit’;

if _n_=m;

Page 380: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 367

proc print data=agreement noobs label;var p_pos lower_p upper_p;

proc print data=agreement noobs label;var p_neg lower_n upper_n;run;

Output from Program 13.2

Proportion Lower 95% Upper 95%of positive confidence confidenceagreement limit limit

0.714 0.446 0.983

Proportion Lower 95% Upper 95%of negative confidence confidenceagreement limit limit

0.913 0.828 0.998

It follows from Output 13.2 that in the schizophrenia diagnosis example we observed71.4% agreement on positive ratings and 91.3% agreement on negative ratings. Using theGraham-Bull approach with these data, we compute the following 95% confidence intervals:(0.446, 0.983) for agreement on positive ratings, and (0.828, 0.998) for agreement onnegative ratings.

13.2.4 Inter-Rater Reliability: Multiple RatersFleiss (1971) proposed a generalized κ statistic for situations with multiple (more thantwo) raters for a group of subjects. It follows the form of κ above, where percent agreementis now the probability that a randomly chosen pair of raters will give the same diagnosis orrating.

EXAMPLE: Psychiatric diagnosis exampleSandifer et al. (1968) provided data from a study in which six psychiatrists provideddiagnoses for each of 30 subjects. Subjects were diagnosed with either depression,personality disorder, schizophrenia, neurosis, or classified as not meeting any of thediagnoses.

Program 13.3 computes the expected agreement (pc), percent agreement (pa), kappastatistic (κ), and a large sample test statistic for testing the null hypothesis of noagreement beyond chance in the psychiatric diagnosis example. These statistics werecalculated using the formulas provided by Fleiss (1971):

pc =∑

i

p2i , pa =

1Nr(r − 1)

⎡⎣∑ij

n2ij − Nr

⎤⎦ , κ =pa − pc

1 − pc,

Var(κ) =2

Nr(r − 1)

∑i p

2i − (2r − 3) [

∑i p

2i ]

2 + 2(r − 2)∑

i p3i

[1 −∑

i p2i ]

2 ,

where N is the total number of subjects, r is the number of raters, and the subscripts i andj refer to the subject and diagnostic category, respectively.

Page 381: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

368 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 13.3 Computation of agreement measures

data diagnosis;input patient depr pers schz neur othr @@;datalines;

1 0 0 0 6 0 2 0 3 0 0 3 3 0 1 4 0 1 4 0 0 0 0 6 5 0 3 0 3 06 2 0 4 0 0 7 0 0 4 0 2 8 2 0 3 1 0 9 2 0 0 4 0 10 0 0 0 0 611 1 0 0 5 0 12 1 1 0 4 0 13 0 3 3 0 0 14 1 0 0 5 0 15 0 2 0 3 116 0 0 5 0 1 17 3 0 0 1 2 18 5 1 0 0 0 19 0 2 0 4 0 20 1 0 2 0 321 0 0 0 0 6 22 0 1 0 5 0 23 0 2 0 1 3 24 2 0 0 4 0 25 1 0 0 4 126 0 5 0 1 0 27 4 0 0 0 2 28 0 2 0 4 0 29 1 0 5 0 0 30 0 0 0 0 6;%let nr=6; * Number of raters;data agreement;

set diagnosis nobs=m;format pa pc kappa kappa_lower kappa_upper 5.3;sm=depr+pers+schz+neur+othr;sum0+depr; sum1+pers; sum2+schz; sum3+neur; sum4+othr;pi=((depr**2)+(pers**2)+(schz**2)+(neur**2)+(othr**2)-sm)/(sm*(sm-1));tot+((depr**2)+(pers**2)+(schz**2)+(neur**2)+(othr**2));smtot+sm;if _n_=m then do;

avgsum=smtot/m;n=sum0+sum1+sum2+sum3+sum4;pdepr=sum0/n; ppers=sum1/n; pschz=sum2/n; pneur=sum3/n; pothr=sum4/n;* Percent agreement;pa=(tot-(m*avgsum))/(m*avgsum*(avgsum-1));* Expected agreement;pc=(pdepr**2)+(ppers**2)+(pschz**2)+(pneur**2)+(pothr**2);pc3=(pdepr**3)+(ppers**3)+(pschz**3)+(pneur**3)+(pothr**3);* Kappa statistic;kappa=(pa-pc)/(1-pc);

end;* Variance of the kappa statistic;kappa_var=(2/(n*&nr*(&nr-1)))*(pc-(2*&nr-3)*(pc**2)+2*(&nr-2)*pc3)/((1-pc)**2);* 95% confidence limits for the kappa statistic;kappa_lower=kappa-1.96*sqrt(kappa_var);kappa_upper=kappa+1.96*sqrt(kappa_var);label pa=’Percent agreement’

pc=’Expected agreement’kappa=’Kappa’kappa_lower=’Lower 95% confidence limit’kappa_upper=’Upper 95% confidence limit’;

if _n_=m;proc print data=agreement noobs label;

var pa pc;proc print data=agreement noobs label;

var kappa kappa_lower kappa_upper;run;

Page 382: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 369

Output from Program 13.3

Percent Expectedagreement agreement

0.556 0.220

Lower 95% Upper 95%confidence confidence

Kappa limit limit

0.430 0.408 0.452

Output 13.3 lists the percent agreement (pa = 0.556), expected agreement (pc = 0.22),and kappa (κ = 0.43). It also shows the 95% confidence limits for κ. The lower limit is wellabove 0 and thus there is strong evidence that agreement in the psychiatric diagnosisexample is in excess of chance. As there is evidence for agreement beyond chance, as afollow-up analysis one may be interested in assessing agreement for specific diagnoses. Fleiss(1971) describes such analyses—computing the conditional probability that a randomlyselected rater assigns a patient with diagnosis j, given that a first randomly selected raterchose diagnosis j. This is analogous to the use of p+ described in the previous section.

13.2.5 Inter-Rater Reliability: Rating Scale DataThe inter-rater reliability of a rating scale is a measure of the consistency of ratings fromdifferent raters at the same point in time. The intra-class correlation coefficient (ICC) orthe concordance correlation coefficient (CCC) are the recommended statistics forquantifying inter-rater reliability when using continuous measures. Shrout et al. (1979)presented multiple forms of the ICC along with discussions of when to use various forms.Here we present the statistics for estimating ICCs from a two-way ANOVA model:

Model outcome = Subjects + Raters.

We will consider the case when randomly selected raters rate each subject (ICC1) andalso the case when the selected raters are the only ones of interest (ICC2). ICC1 may be ofinterest when it can be assumed the raters in a study represent a random sample of apopulation for which the researchers would like to generalize to. ICC2 may be used in ratertraining sessions prior to a clinical trial with pre-selected raters. The two coefficients aredefined as follows:

ICC1 =Ms − Me

Ms + (r − 1)Me + r(Mr − Me)/n,

ICC2 =Ms − Me

Ms + (r − 1)Me,

where r is the number of raters, n is the number of subjects, and Ms, Mr, and Me are themean squares for subjects, raters, and error, respectively.

Shrout et al. (1979) also provides formulas for estimating the variances for each form ofthe ICC and a discussion of a one-way ICC when different sets of raters score each subject.The null hypothesis of a zero ICC can be tested with an F -test given by F = Ms/Me with(n − 1) and (r − 1)(n − 1) degrees of freedom. As the ICC statistic and the associated testsand confidence intervals are based on summary statistics produced in a typical ANOVA,these can be easily computed using either the GLM or MIXED procedure. The SAS libraryof sample programs provides a macro that computes all versions of the ICC per Shrout’sformulas as well as others:

Page 383: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

370 Pharmaceutical Statistics Using SAS: A Practical Guide

http://ftp.sas.com/techsup/download/stat/intracc.html

Further discussion of the ICC and related statistics is provided later in Section 13.2.7.

EXAMPLE: Rater training session exampleWe will now describe an example of a rater training session performed at the beginning ofa clinical trial. While in the previous examples a small number of raters assessed manysubjects, in this example many raters assess only a small number of subjects (often one ortwo). This situation is more commonplace in clinical trials. That is, a rater training sessionis a good example of this setting.

Prior to implementing an antidepressant clinical trial, Demitrack et al. (1998) assessedthe inter-rater reliability of 85 raters for the Hamilton Rating Scale for Depression(HAMD) (Hamilton, 1960) as part of a rater training session. The HAMD total score is ameasure of the severity of a patient’s depression with higher scores indicating more severedepression. The main goal of the session was to establish consistency in the administrationand interpretation of the scale across multiple investigational sites. From a statisticalperspective, the goals were to reduce variability (and thus increase power) by trainingraters to score using the scale in a similar fashion and to identify potential outlier ratersprior to the study.

Raters independently scored the HAMD from a videotaped interview of a depressedpatient. Fleiss’ generalized κ, as described in the previous section, was used to summarizethe inter-rater reliability for this group of raters. As only one subject was assessed,agreement was computed across items of a scale rather than across subjects. The percentagreement now represents the probability that two randomly selected raters provided thesame rating score for a randomly selected item. The observed agreement and κ for thesedata are 0.55 and 0.4.

While an overall summary of agreement provides information about the scale and raters,it is also of importance in a multi-site clinical trial to identify raters who are outliers.These raters have the potential to increase variability in the outcome variable and thusreduce the power to detect treatment differences. Using the mode of the group of raters asthe gold standard, Demitrack et al. (1998) computed the percent agreement and inter-classcorrelation coefficient for each rater (relative to the gold standard). Rater ICCs rangedfrom 0.40 to 1.0, with only two raters scoring below 0.70. Percentage agreement was alsoassessed by item in order to identify areas of disagreement for follow-up discussion.

Rater training sessions followed the discussion of the initial videotape results, and thesession concluded with raters scoring a videotape of the same patient in an improvedcondition. Demitrack et al. concluded that the training did not clearly improve theagreement of the group, though they were able to use the summary agreement statistics toidentify raters who did not score the HAMD consistent with the remainder of the groupprior to implementing the trial. While Demitrack et al. used percent agreement and ICC intheir work, the average squared deviation (from the gold standard) is another statistic thathas been proposed for use in summarizing inter-rater reliability data (Channon and Butler,1998).

13.2.6 Internal ConsistencyMeasures of internal consistency assess the degree to which each item of a rating scalemeasures the same construct. Cronbach’s α, a measure of the average correlation of itemswithin a scale, is a commonly used statistic to assess internal consistency (Cronbach, 1951):

α =k

k − 1

[1 −

∑i Vi

V

],

Page 384: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 371

where k is the number of items, Vi is the variance of the ith item, and V is the variance ofthe total score.

Cronbach’s α ranges from 0 to 1, with higher scores suggesting good internalconsistency. It is considered to be a lower bound of the scale’s reliability (Shrout, 1998).Perrin suggested 0.7 as a minimal standard for internal consistency when making groupcomparisons, with a higher standard of at least 0.90 when individual comparisons are to beused (Perrin et al., 1997). Cronbach’s α is a function of the average inter-item correlationand the number of items. It increases as the number of related items in the scale increases.If α is very low, the scale is either too short or the items have very little in common (deleteitems which do not correlate with the others). On the other hand, extremely high scoresmay indicate some redundancy in the rating scale. Cronbach’s α can be computed usingthe CORR procedure.

EXAMPLE: EESC rating scale exampleKratochvil et al. (2004) reported on the development of a new rating scale with 29 items toquantify expression and emotion in children (EESC). Data from an interim analysis of avalidation study involving 99 parents of children with attention-deficit hyperactivitydisorder are used here to illustrate the assessment of internal consistency.

Program 13.4 uses PROC CORR for computing internal consistency and item-to-totalcorrelations in the EESC rating scale example. The EESC data set used in the programcan be found on the book’s companion Web site.

Program 13.4 Computation of Cronbach’s α

proc corr data=eesc alpha;var eesc1 eesc2 eesc3 eesc4 eesc5 eesc6 eesc7 eesc8 eesc9 eesc10

eesc11 eesc12 eesc13 eesc14 eesc15 eesc16 eesc17 eesc18 eesc19 eesc20eesc21 eesc22 eesc23 eesc24 eesc25 eesc26 eesc27 eesc28 eesc29;

run;

Output from Program 13.4

Cronbach Coefficient Alpha

Variables Alpha

Raw 0.900146Standardized 0.906284

Cronbach Coefficient Alpha with Deleted Variable

Raw Variables Standardized Variables

Deleted Correlation CorrelationVariable with Total Alpha with Total Alpha

EESC1 0.291580 0.899585 0.281775 0.906622EESC2 0.545988 0.895571 0.564959 0.901673EESC3 0.591867 0.894167 0.593887 0.901158EESC4 0.672391 0.892861 0.672410 0.899748EESC5 0.247457 0.901908 0.240618 0.907326EESC6 0.458779 0.896995 0.448555 0.903730EESC7 0.478879 0.896641 0.472999 0.903300

Page 385: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

372 Pharmaceutical Statistics Using SAS: A Practical Guide

EESC8 0.665566 0.893793 0.675876 0.899685EESC9 0.505510 0.896072 0.498409 0.902853EESC10 0.651696 0.894064 0.652332 0.900110EESC11 0.666862 0.893457 0.677098 0.899663EESC12 0.380065 0.898422 0.395765 0.904652EESC13 0.368793 0.898586 0.363621 0.905211EESC14 0.623236 0.893495 0.614713 0.900785EESC15 0.392515 0.898165 0.407936 0.904440EESC16 0.389488 0.898200 0.396298 0.904643EESC17 0.624384 0.894600 0.637290 0.900380EESC18 0.485442 0.896484 0.480872 0.903162EESC19 -.015219 0.907669 -.000957 0.911384EESC20 0.707713 0.891929 0.710875 0.899052EESC21 0.550139 0.895331 0.540735 0.902104EESC22 0.639571 0.894357 0.649545 0.900160EESC23 0.436383 0.897693 0.435663 0.903956EESC24 0.423187 0.897806 0.424149 0.904157EESC25 0.294496 0.901074 0.292439 0.906439EESC26 0.313600 0.899842 0.315250 0.906047EESC27 0.514513 0.896137 0.512612 0.902602EESC28 0.362475 0.898664 0.367987 0.905135EESC29 0.509206 0.896090 0.508525 0.902674

Output 13.4 provides estimates of Cronbach’s α using both raw data and itemsstandardized to have a standard deviation of 1.0. The standardized scores may be usefulwhen there are large differences in item variances. The results indicate an acceptable levelof internal consistency (α > 0.90).

The output also lists the correlation of each item to the remaining total score, as thevalue of Cronbach’s α with each individual item deleted from the scale, as well as thecorrelation of each item score with the remaining total score. The item-to-total correlationswere at least moderate for all items except number 19. The low item-to-total for item 19 ledto further investigation. It was found that one item (“My child shows a range of emotions”)was being interpreted in both a positive and negative manner by different parents, leadingto the poor correlation. Thus, this item was removed in the revised version of the scale.

Despite the acceptable properties, it may still be of interest to consider a more concisescale, one that would be less burdensome on the respondent. From Output 13.3, theremoval of any single item would not bring about a substantial change in Cronbach’s α.Inter-item correlations (also produced by Program 13.3 but not included in the output tosave space) could be examined to make scale improvements in this situation.

The PROC CORR documentation contains another example of the computation ofCronbach’s α.

13.2.7 Test-Retest ReliabilityTest-retest reliability indicates the stability of ratings on the same individual at twodifferent times while in the same condition. For test-retest assessments, the retest ratingsshould be taken long enough after the original assessment to avoid recall bias and soonenough to limit the possibility of changes in the condition of the subject. For continuousmeasures, Pearson’s correlation coefficient is often used to quantify the relationshipbetween two measures. However, as this captures only the correlation between measures,and not whether the measures are systematically different, the ICC or CCC are preferredmeasures for quantifying test-retest reliability. For instance, if the second (retest)measurement is always 2 points higher than the first, Pearson’s correlation coefficient willbe 1 while the ICC and CCC will be reduced due to the systematic differences. Consideringthese data as a scatterplot with the x and y axis representing the first and second

Page 386: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 373

measurements on the subjects, Pearson’s correlation coefficient assesses deviations from thebest fitting regression line. The CCC is a measure that assesses departures from the line ofidentity (a 45 degree line). Thus, systematic departures will reduce the CCC. The closelyrelated ICC statistic is the proportion of the total variability in scores that is due to thevariability among subjects. Thus, an ICC close to 1 indicates that little of the variability inscores is due to rater differences, and that thus the test-retest reliability is excellent.

The (2-way) ICC and CCC statistics are given by (Shrout and Fleiss, 1979; Lin et al.,2002)

ICC =Ms − Me

Ms + (m − 1)Me, CCC =

2rs1s2

s21 + s2

2 + (y1 + y2)2 ,

where Ms and Me are the mean squares for subjects and error, respectively, r is Pearson’scorrelation coefficient between observations at the first and second time points, i.e., Yi1 andYi2, and s2

1 and s22 are the sample variances of the outcomes at the first and second time

points. The 95% confidence limits for ICC are given below(FL − 1

FL + k − 1,

FU − 1FU + k − 1

),

where k is the number of occasions,

FL = F0/F1−α/2[n − 1, (n − 1)(k − 1)],

FU = F0F1−α/2[(n − 1)(k − 1), n − 1]

and F0 = Ms/Me.Lin (1989) demonstrated that

Z =12

ln1 − CCC

1 + CCC

is asymptotically normal with mean

12

ln1 − C

1 + C

and variance

1n − 2

[(1 − r2)C2

r2(1 − C2)+

4C3(1 − C)μ2

r(1 − C2)2 − 2C4μ4

r2(1 − C2)2

],

where C is the true concordance correlation coefficient, r is Pearson’s correlation coefficientdefined above, and μ = (μ1 − μ2)/

√σ1σ2. Confidence intervals can be formed by replacing

the parameters with their estimates.An alternative testing approach to assessing inter-rater reliability is provided by the

Bradley-Blackwood test. This approach simultaneously tests the null hypothesis of equalmeans and variances at the first and second assessments (Sanchez and Binkowitz, 1999).The Bradley-Blackwood test statistic B, which has an F distribution with 2 and n − 2degrees of freedom, is as follows:

B =1

2Mr

[∑i

D2i − Sr

],

where Di = Yi1 − Yi2, and Sr and Mr are the error sum of squares and mean square error ofthe regression of Di on Ai = (Yi1 + Yi2)/2.

Page 387: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

374 Pharmaceutical Statistics Using SAS: A Practical Guide

EXAMPLE: Sickness inventory profile exampleDeyo et al. (1991) provided data on the sickness inventory profile (SIP) from a clinical trialfor chronic lower back pain. Data were obtained from 34 patients whose symptoms weredeemed unchanged in severity, with measures taken two weeks apart.

Program 13.5 computes the ICC and associated large sample 95% confidence limits. Asthe ICC is computed from mean squares in an ANOVA model, it is easily computable usingoutput from PROC MIXED as demonstrated below.

Program 13.5 Computation of the ICC, CCC and Bradley-Blackwood test

options ls=70;data inventory;

input patient time sip @@;datalines;

1 1 9.5 9 1 7.3 17 1 4.0 25 1 10.1 33 1 4.61 2 12.3 9 2 6.1 17 2 3.6 25 2 15.7 33 2 4.32 1 3.6 10 1 0.5 18 1 6.6 26 1 10.2 34 1 4.62 2 0.4 10 2 1.0 18 2 7.0 26 2 15.2 34 2 4.33 1 17.4 11 1 6.0 19 1 13.8 27 1 9.53 2 7.4 11 2 3.1 19 2 10.1 27 2 8.24 1 3.3 12 1 31.6 20 1 9.8 28 1 19.14 2 2.2 12 2 16.6 20 2 8.3 28 2 21.95 1 13.4 13 1 0 21 1 4.8 29 1 5.65 2 6.0 13 2 2.3 21 2 2.9 29 2 10.66 1 4.1 14 1 25.0 22 1 0.9 30 1 10.76 2 3.5 14 2 12.2 22 2 0.4 30 2 13.17 1 9.9 15 1 3.4 23 1 8.0 31 1 8.67 2 9.9 15 2 0.7 23 2 2.8 31 2 6.18 1 11.3 16 1 2.8 24 1 2.7 32 1 7.58 2 10.6 16 2 1.7 24 2 3.8 32 2 5.2

;* Intraclass correlation coefficient;proc sort data=inventory;

by patient time;proc mixed data=inventory method=type3;

class patient time;model sip=patient time;ods output type3=mstat1;

data mstat2;set mstat1;dumm=1;if source=’Residual’ then do; mserr=ms; dferr=df; end;if source=’patient’ then do; mspat=ms; dfpat=df; end;if source=’time’ then do; mstime=ms; dftime=df; end;retain mserr dferr mspat dfpat mstime dftime;keep mserr dferr mspat dfpat mstime dftime dumm;

data mstat3;set mstat2;by dumm;format icc lower upper 5.3;if last.dumm;icc=(mspat-mserr)/(mspat+(dftime*mserr));fl=(mspat/mserr)/finv(0.975,dfpat,dfpat*dftime);fu=(mspat/mserr)*finv(0.975,dfpat*dftime,dfpat);lower=(fl-1)/(fl+dftime);upper=(fu-1)/(fu+dftime);

Page 388: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 375

label icc=’ICC’lower=’Lower 95% confidence limit’upper=’Upper 95% confidence limit’;

proc print data=mstat3 noobs label;var icc lower upper;run;

* Concordance correlation coefficient;data transpose;

set inventory;by patient;retain base;if first.patient=1 then base=sip;if last.patient=1 then post=sip;diff=base-post;if last.patient=1;

proc means data=transpose noprint;var base post diff;output out=cccout mean=mn_base mn_post mn_diff var=var_base var_post var_diff;

data cccout;set cccout;format ccc 5.3;ccc=(var_base+var_post-var_diff)/(var_base+var_post+mn_diff**2);label ccc=’CCC’;

proc print data=cccout noobs label;var ccc;run;

* Bradley-Blackwood test;data bb;

set transpose;avg=(base+post)/2; difsq=diff**2; sdifsq+difsq;dum=1;run;

proc sort data=bb;by dum;

data bbs;set bb;by dum;if last.dum;keep sdifsq;

proc mixed data=bb method=type3;model diff=avg;ods output type3=bb1;

data bb1;set bb1;if source=’Residual’;mse=ms; sse=ss; dfe=df;keep mse sse dfe;

data test;merge bb1 bbs;format f p 5.3;f=0.5*(sdifsq-sse)/mse;p=1-probf(f,2,dfe+2);label f=’F statistic’

p=’P-value’;proc print data=test noobs label;

var f p;run;

Page 389: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

376 Pharmaceutical Statistics Using SAS: A Practical Guide

Output from Program 13.5

Lower 95% Upper 95%confidence confidence

ICC limit limit

0.727 0.520 0.854

CCC

0.706

Fstatistic P-value

4.194 0.024

Output 13.5 shows that the ICC for the data collected in the sickness inventory profilestudy is 0.727, with a 95% confidence interval of (0.520, 0.854). Landis and Koch (1977)indicate that ICCs above 0.60 suggest satisfactory stability, and ICCs greater than 0.80correspond to excellent stability. For reference, the CCC based on these data is 0.706, verysimilar to the ICC. See Deyo (1991) for reformulations of the formula for computing theCCC which shows the similarity between the CCC and ICC.

In addition, Output 13.5 lists the F statistic and p-value of the Bradley-Blackwood test.The test rejects the null hypothesis of equal means and variances (F = 4.194, p = 0.024).Thus, the Bradley-Blackwood test would seem to be contradictory to the relatively highICC value. However, one must consider that the various statistics are differentially sensitiveto the strength of the linear relationship, location, and scale shifts between the first andsecond measurement. Sanchez et al. (1999) discussed these issues in depth and performed asimulation study with multiple reliability measures. They conclude that the CCC comesthe closest among all the measures to satisfying all the components of a good test-retestreliability measure. Regardless, they recommend computing estimates of scale and locationshifts along with the CCC.

13.3 Validity and Other TopicsA measure is said to be valid if it accurately reflects the concept it is intended to measure.Assessing the validity of a measure typically includes quantifying convergent validity,divergent validity, and discriminant validity. These three topics, which comprise theassessment of the construct validity of an instrument, can be addressed in a quantitativefashion and will be discussed below.

Content validity is another topic which is typically addressed in the development stageof a measurement tool. Content validity refers to the degree to which a measure covers therange of meanings that are part of the concept to be measured. The most commonapproaches to assessing content validity are expert reviews of the clarity,comprehensiveness, and redundancy of the measurement tool. Content validity will not bediscussed further in this chapter.

However, responsiveness, factor analysis, and minimal clinically relevant differences arethree additional topics that are discussed. Responsiveness and factor structure of ameasurement tool are two important practical concepts to assess when validating aninstrument. Establishing a minimal clinically relevant difference addresses a common needfor applied researchers designing studies using and interpreting data from studies with ameasurement tool.

Page 390: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 377

13.3.1 Convergent and Divergent ValidityConvergent validity is established by showing a strong relationship between the scale underreview and another validated scale thought to measure the same construct. Divergentvalidity is the lack of correlation between the scale under review and scales thought toassess different constructs. Pearson’s correlation coefficient is the most commonly usedstatistic to quantify convergent and divergent validity. Thus, SAS can be easily used tosummarize convergent and divergent validity through the CORR procedure. A Pearson’scorrelation of at least 0.4 has been used (Cappelleri et al., 2004) as evidence for convergentvalidity. Similarly, correlations less than 0.3 indicate evidence for divergent validity, withcorrelations between 0.3 and 0.4 taken as no evidence to establish or dismiss convergent ordivergent validity. Confidence intervals are useful here to provide information beyondsimple point estimates.

Cappelleri et al. (2004) provides an example of the assessment of convergent anddivergent validity in assessing a new scale for self-esteem and relationships (SEAR) inpatients with erectile dysfunction. As part of the assessment of convergent/divergentvalidity, they included a global quality of life scale (SF-36) in a validation study. Theyhypothesized low correlations between the Confidence domain of the SEAR with physicalfactors of the SF-36 and thus used these comparisons to assess divergent validity. Similarlyfor convergent validity, the developers hypothesized higher correlations with SF-36 mentalhealth domains. Table 13.3 presents the results.

Table 13.3 Correlation of SF-36 Components withSEAR Confidence Domain

SF-36 component Correlation

Physical Functioning 0.30Role-Physical 0.30Bodily Pain 0.32

Mental Health 0.44Role-Emotional 0.45Mental Component Summary 0.45

The correlations greater than 0.4 with the mental health measures were taken asevidence for convergent validity while the correlations with physical domains wereborderline evidence (at least no evidence to dismiss) for divergent validity.

13.3.2 Discriminant ValidityDiscriminant validity indicates the ability of a scale to distinguish different groups ofsubjects. An instrument for assessing the severity of a disease should clearly be able todistinguish between subjects with and without the corresponding diagnosis. Often,however, many disease symptoms overlap with other diseases, such as patients diagnosedwith depression versus anxiety disorder. Thus in establishing validity of a measure for aparticular disease, ability to distinguish between a disease state with overlapping symptomsmay be extremely important. Note that there is some inconsistency in the literatureregarding the use of the term discriminant validity—with references defining this in thesame meaning as divergent validity above (for example, Guyatt and Feeny, 1991). However,in this discussion, discriminant validity is defined as the ability to distinguish betweenrelevant subject groups.

Thurber et al. (2002) assessed the discriminant validity of the Zung self-ratingdepression scale in 259 individuals referred to a state vocational rehabilitation service. TheZung scale was administered as part of a test battery, including the depression subscale ofthe Minnesota Multiphasic Personality Inventory-2 (MMPI-2), following a diagnostic

Page 391: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

378 Pharmaceutical Statistics Using SAS: A Practical Guide

interview. Using logistic regression and stepwise logistic regression (such as the LOGISTICprocedure), they demonstrated that the Zung scale was a predictor (the strongest) ofdepressed versus non-depressed individuals, as well as between individuals diagnosed withdepression or substance abuse. The Zung scale was also predictive of a diagnosis ofdepression even after the forced inclusion of the MMPI-2 depression subscale into themodel.

Once the scale has been demonstrated to be predictive of a diagnosis, the ability of thescale to predict the diagnosis for individual subject is often assessed using receiver operatorcharacteristic (ROC) curves. The ROC is a graph of sensitivity versus (1-specificity) forvarious cutoff scores. For these data, based on a ROC curve, Thurber chose a cutoff scoreof 60 on the Zung scale for identifying subjects with and without a diagnosis of depression.With this cutoff score, sensitivity (the proportion of depressed subjects correctly identified)was 0.57, while specificity (the proportion of subjects without depression who werecorrectly identified) was 0.83. See Deyo et al. (1991) for a further discussion of ROC and anexample using changes in SIP scores (see Section 13.2.7) to predict improvement status.

13.3.3 ResponsivenessResponsiveness is the ability of an instrument to detect small but clinically importantchanges. Responsiveness is often referred to as sensitivity to change and is often viewed aspart of construct validity. Because the main purpose of a clinical trial is to detect atreatment effect, it is important to assess the responsiveness of a scale prior to using it in aclinical trial. A scale that is not responsive may not be able to detect important treatmentchanges and therefore mislead the experimenter to conclude no treatment effect. Themethods discussed above are predominantly point in time analyses (except for test-retestreliability which focuses on stability of scores) and do not fully demonstrate that aninstrument would be effective for a clinical trial designed to detect differences in changescores. The standardized response mean (the mean change divided by the standarddeviation in change scores) is a common unitless statistic for summarizing responsiveness(Stratford et al., 1996). Change scores may be summarized following an interventionexpected to produce a clinically relevant change. When a control is available, effect sizes foran effective intervention relative to the control may be compared using multiple measures.

For example, Faries et al. (2001) assessed the responsiveness of the ADHD rating scalewhen administered by trained clinicians. The scale had been previously validated as aparent scored tool. As no control group was available, the SRM was used to compare thechanges observed on the new version of the scale with other validated instruments. Resultsshowed the observed SRM for the clinician scored scale (1.21) was in the range of SRMsobserved from other clinician and parent measures (1.13 to 1.40). As the SRM is based onsimple summary statistics, PROC UNIVARIATE provided the information necessary forthe computation of the SRM.

Responsiveness of an instrument may also be assessed using correlations with a globalimprovement scale. The global improvement question usually asks the subjects to rate theimprovement on their condition on an ordinal scale. An example of such scale would be“very much better”, “much better”, “a little better”, “no change”, “a little worse”, “muchworse” and “very much worse”. Mean changes in the instrument scores are calculated forsubjects in each global scale response category regardless of the treatment. A monotoneincreasing (or decreasing, depending on the direction of improvement) function of the meanscores is a desirable property for a scale that is responsive to treatment. An analysis ofvariance (ANOVA) can be performed to test the mean differences in the mean instrumentscores among subjects in different global scale response categories. The model shouldinclude the change in the instrument scores as the dependent variables and the global scaleresponse categories as the class variable. A responsive scale should be able to discriminatereasonably well between the response categories of a global rating scale.

Page 392: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 379

13.3.4 Identifying SubscalesIt is often desired to identify subscales of a multi-item questionnaire in addition to itsoverall total score. Factor analysis is a technique commonly used to explore the existence ofsuch subscales. It describes the covariance relationships among many items in terms of afew underlying, but unobservable variables called factors. Each factor may correspond to asubscale. If items can be grouped by their correlations and all items within a particulargroup are highly correlated among themselves but have relatively small correlations withitems in a different group, it is feasible that each group of items represents a singleunderlying construct, or a subscale.

One approach is to come up with an a priori subscale structure proposed by the expertsin that field and test the appropriateness of that structure by using the confirmatory factoranalysis. In this case, the experts should identify the subscales first and allocate the itemsto each subscale based on their opinion. Then a confirmatory factor analysis model can beused to test the fit of the model. In a confirmatory factor analysis model, the researchermust have an idea about the number of factors and know which items load on whichfactors. The model parameters are defined accordingly prior to fitting the model. Forexample if an item is hypothesized to load on a specific factor, the corresponding factorloading will be estimated and the loadings corresponding to this item on the other factorswill be set to zero. After estimating the parameters, the fit of this model is tested to assessthe appropriateness of the model. Fitting a confirmatory factor analysis model requiressome more detailed knowledge about factor analysis. Details about confirmatory factoranalysis and using the CALIS procedure to fit the model can be found in Hatcher (1994).

Another way is to use an exploratory factor analysis to identify the number of subscalesand the items that will be allocated to each subscale. As opposed to the confirmatoryfactor analysis, the researcher is usually unsure about the number of factors and how theitems will load on the factors. After identifying the subscales, the experts should approvethe face validity of the subscales. Face validity is not validity in technical sense. It is notconcerned with what the test actually measures, but what it appears superficially tomeasure. For example, a psychiatrist could review the items loaded on a “Sleep Problems”subscale of a depression symptom questionnaire to see if any of those items appear to berelated to measuring sleep disturbances on a depressed patient.

Exploratory factor analysis usually involves two stages. The first is to identify thenumber of factors and estimate the model parameters. There are several methods ofestimation of the model parameters. The most commonly used are the PrincipalComponent and Maximum Likelihood methods. As initial factors are typically difficult tointerpret, a second stage of rotation makes the final result more interpretable. A factorrotation is carried out to look for a pattern of loadings such that each item loads highly ona single factor (subscale) and has small to moderate loadings on the remaining factors.This is called simple structure. Orthogonal and oblique factor rotations are two types oftransformations may be needed to achieve the simple structure. Orthogonal rotations areappropriate for a factor model in which the common factors are assumed to beuncorrelated and oblique to be correlated. The type of transformation (orthogonal versusoblique) can be decided by using a graphical examination of factor loadings (whether arigid or nonrigid transformation make the factor loadings close to the axes). For detailedinformation about factor analysis, see Johnson and Wichern (1982) and Hatcher (1994).

EXAMPLE: Incontinence Quality of Life questionnaireIncontinence Quality of Life questionnaire (I-QOL) is a validated incontinence-specificquality of life questionnaire that includes 22 items (Wagner et al., 1996; Patrick et al.,1999). The I-QOL yields a total score and three subscale scores: Avoidance and LimitingBehaviors, Psychosocial Impacts, and Social Embarrassment. For simplicity, we selectednine of the items in two subscales to demonstrate how an exploratory factor analysis can be

Page 393: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

380 Pharmaceutical Statistics Using SAS: A Practical Guide

used to define two of the three subscales. The data were obtained from a pre-randomizationvisit of a pharmacotherapy clinical trial for treatment of women with urinary incontinence.

Program 13.6 uses the FACTOR procedure to perform an exploratory factor analysis ofthe I-QOL data (the IQOL data set used in the program can be found on the book’scompanion Web site). PROC FACTOR can be used to fit a linear factor model andestimate the factor loadings. The SCREE option plots the eigenvalues associated with eachfactor to help identify the number of factors. An eigenvalue represents the amount ofvariance that is accounted for by a given factor. The scree test looks for a break orseparation between the factors with relatively large eigenvalues and those with smallereigenvalues. The factors that appear before the break are assumed to be meaningful andare retained. Figure 13.1 produced by Program 13.6 indicates that two factors weresufficient to explain most of the variability. Although the number of factors can be specifiedin PROC FACTOR for the initial model, this number is just an intuitive feeling and it canbe changed in subsequent analyses based on empirical results. Even though the NFACToption specifies a certain number of factors, the output will still include the scree plot andthe eigenvalues, with the number of factors extracted being equal to the number ofvariables analyzed. However, the parts of the output related to factor loadings and therotations will include only the number of factors specified with the NFACT option.

The ROTATE and REORDER options are used to help interpret the obtained factors.In order to achieve the simple structure, the VARIMAX rotation was carried out in thisexample. The REORDER option reorders the variables according to their largest factorloadings. The SIMPLE option displays means, standard deviations, and the number ofobservations. Upon examination of the scree plots, eigenvalues, and the related factors, thenumber of factors in the NFACT option should be changed to the appropriate level, andthen the model should be re-run. This examination may include the scree plot, eigenvalues,and the rotated factors together. In some instances, it could be desirable to keep factorswith eigenvalues less than 1, if interpretation of these factors makes sense. The maximumlikelihood method (METHOD=ML) is useful since it provides a chi-square test for modelfit. However, as the test is a function of the sample size, for large studies the test mayreject the hypothesis of sufficient number of factors due to differences that are not clinicallyrelevant. Therefore it is recommended to use other goodness-of-fit indices (Hatcher, 1994).

Program 13.6 Subscale identification in the I-QOL example

proc factor data=iqol simple method=ml scree heywood reorder rotate=varimax nfact=2;var iqol1 iqol4 iqol5 iqol6 iqol7 iqol10 iqol11 iqol17 iqol20;ods output Eigenvalues=scree;

data scree;set scree;if _n_<=9;number=_n_;

* Vertical axis;axis1 minor=none label=(angle=90 "Eigenvalue") order=(-1 to 11 by 2);* Horizontal axis;axis2 minor=none label=("Number") order=(1 to 9);symbol1 i=none value=dot color=black height=3;proc gplot data=scree;

plot eigenvalue*number/vaxis=axis1 haxis=axis2 vref=0 lvref=34 frame;run;quit;

Page 394: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 381

Figure 13.1 Scree plot in the I-QOL example

Eig

enva

lue

-1

1

3

5

7

9

11

Number

1 2 3 4 5 6 7 8 9

Output from Program 13.6

Initial Factor Method: Maximum Likelihood

Factor Pattern

Factor1 Factor2

IQOL6 IQOL item 6 0.79447 0.10125IQOL11 IQOL item 11 0.77077 -0.16251IQOL17 IQOL item 17 0.76174 0.39284IQOL7 IQOL item 7 0.74458 0.38372IQOL4 IQOL item 4 0.69355 -0.46179IQOL5 IQOL item 5 0.69353 0.19886IQOL20 IQOL item 20 0.65887 -0.20804IQOL1 IQOL item 1 0.64754 -0.37380IQOL10 IQOL item 10 0.64487 -0.30917

Rotated Factor Pattern

Factor1 Factor2

IQOL4 IQOL item 4 0.80996 0.19553IQOL1 IQOL item 1 0.71412 0.22151IQOL10 IQOL item 10 0.66487 0.26344IQOL11 IQOL item 11 0.64271 0.45545IQOL20 IQOL item 20 0.60014 0.34238IQOL17 IQOL item 17 0.22891 0.82594IQOL7 IQOL item 7 0.22394 0.80715IQOL6 IQOL item 6 0.46518 0.65195IQOL5 IQOL item 5 0.32498 0.64414

Output 13.6 displays selected sections of the output produced by Program 13.6. The topportion of the output (under “Initial Factor Method: Maximum Likelihood”) presents the

Page 395: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

382 Pharmaceutical Statistics Using SAS: A Practical Guide

factor loadings after fitting the model with two factors (NFACT=2). All items have highloadings on the first factor and small loadings on the second, suggesting a rotation (eachfactor retained should represent some of the items). The bottom portion of Output 13.6(under “Rotated Factor Pattern”) displays the factor loadings after the VARIMAXrotation. The transformed factor loading structure suggests that the first factor (subscale)should consist of Items 1, 4, 10, 11 and 20 since those items are heavily loaded on Factor 1.Similarly the second subscale should consist of Items 5, 6, 7, and 17. At this stage, theresearcher should decide if the items that load on a given factor share some conceptualmeaning and if the items that load on different factors seem to be measuring differentconstructs. In this example, the first subscale was called as Avoidance and LimitingBehaviors, and the second one was called Psychosocial Impacts in the original version ofthe I-QOL subscale creation.

13.3.5 Minimal Clinically Important DifferencesAnother important need is to determine the between- and within-treatment minimumclinically important differences (MCID) for an instrument. The MCID helps cliniciansinterpret the relevance of changes in the instrument scores. The within-treatment MCID isdefined as the improvement in a score with treatment at which a patient recognizes thatshe or he is improved. The between-treatment MCID is the minimum difference betweentwo treatments that can be considered clinically relevant. One widely accepted way todetermine the MCIDs is to anchor the scale to a global rating of change scale such as theone mentioned in the responsiveness discussion. The mean change in the measure ofinterest for those subjects who rated as “a little better” could be considered as thewithin-treatment MCID. The difference in the mean changes for subjects who rated as “alittle better” and who rated “no change” could be considered as the between-treatmentMCID. The between-treatment MCID can be a sound choice for the treatment difference inorder to power the clinical studies. These two MCIDs provide guidance to researchers tointerpret the change scores for the instrument. They become critical when statisticallysignificant differences needed to be justified as clinically relevant. The choice of a globalscale is an important step in determining the MCIDs. Global scales with items that are lesssensitive to change may yield larger MCIDs. For example, if the responses to a global scaleof improvement are “better,” “no change,” or “worse,” the MCIDs calculated using thisscale may be larger than those calculated from the global scale in the previous example.Therefore, the differences could still be clinically important but may not necessarily beminimum using this scale.

13.4 SummaryThis chapter summarizes the importance of and methods for establishing the reliability andvalidity of rating scales. Quality quantitative scientific research must include the use ofmeasurement tools that have been validated for the population under study. The validationof an instrument is not a single test, but a summary of multiple psychometric properties.That is, a scale should have acceptable levels of internal consistency, test-retest reliability,inter-rater reliability, convergent, divergent and discriminant validity, as well asresponsiveness.

A rating scale is said to be reliable if multiple measurements on a subject agree. Aspectsof reliability presented in this chapter include internal consistency, test-retest reliability,and inter-rater reliability. A measure is said to be valid if it accurately reflects the conceptit is intended to measure. Validity discussions included in this chapter include convergent,divergent, and discriminant validity, as well as responsiveness and assessment of the factorstructure of the instrument.

Page 396: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 13 Reliability and Validity: Assessing the Psychometric Properties of Rating Scales 383

Throughout the chapter, multiple statistical methods for assessing the various aspects ofreliability and validity are presented. The statistical methods are presented along withexamples from clinical trials, diagnostic studies, rater training sessions, and scale validationstudies. Multiple examples include SAS code and output to facilitate understanding andease of application of these concepts.

ReferencesAgresti, A. (1992). “A Survey of Exact Inference for Contingency Tables.” Statistical Science. 7,

131–177.Bartko, J.J., Carpenter, W.T. (1976). “On the Methods and Theory of Reliability.” The Journal of

Nervous and Mental Disease. 163, 307–317.Bech, P., Allerup, P., Gram, L.F., Reisby, N., Rosenberg, R., Jacobsen, O., Nagy, A. (1981). “The

Hamilton Depression Scale: Evaluation of Objectivity Using Logistic Models.” Acta PsychiatricaScandinavica. 64, 290–299.

Beretvas, S.N., Pastor, D.A. (2003). “Using Mixed-Effects Models in Reliability GeneralizationStudies.” Educational and Psychological Measurement. 63, 75–95.

Cappelleri, J.C., Althof, S.E., Siegel, R.L., Shpilsky, A., Bell, S.S., Duttagupta, S. (2004).“Development and Validation of the Self-Esteem and Relationship (SEAR) Questionnaire inErectile Dysfunction.” International Journal of Impotence Research. 16, 30–38.

Channon E., Butler A. (1998). “Comparing investigators’ use of rating scales such as PANSS inmulti-investigator studies of schizophrenia.” European Neuropsychopharmacology. 8, S310–311.

Cicchetti, D.V., Allison, T. (1971). “A New Procedure for Assessing Reliability of Scoring EEGSleep Recordings.” American Journal of EEG Technology. 11, 101–109.

Cicchetti, D.V., Feinstein, A.R. (1990). “High Agreement But Low Kappa: II. Resolving theParadoxes.” Journal of Clinical Epidemiology. 43, 551–558.

Cronbach, L.J. (1951). “Coefficient alpha and the internal structure of tests.” Psychometrika. 16,297–334.

Demitrack, M.A., Faries, D., Herrera, J.M., DeBrota, D.J., Potter, W.Z. (1998). “The Problem ofMeasurement Error in Multisite Clinical Trials.” Psychopharmacology Bulletin. 34, 19–24.

Deyo, R.A., Diehr, P., Patrick, D.L. (1991). “Reproducibility and Responsiveness of Health StatusMeasures: Statistics and Strategies for Evaluation.” Controlled Clinical Trials. 12, S142–158.

Donner, A., Eliasziw, M. (1987). “Sample Size Requirements for Reliability Studies.” Statistics inMedicine. 6, 441–448.

Evans, K.R., Sills, T., DeBrota, D.J., Gelwicks, S., Engelhardt, N., Santor, D. (2004). “An ItemResponse Analysis of the Hamilton Depression Rating Scale Using Shared Data From TwoPharmaceutical Companies.” Journal of Psychiatric Research. 38, 275–284.

Faries, D.E., Yalcin, I., Harder, D., Heiligenstein, J.H. (2001). “Validation of the ADHD RatingScale as a Clinician Administered and Scored Instrument.” Journal of Attention Disorders. 5,107–115.

Fleiss, J.L. (1971). “Measuring Nominal Scale Agreement Among Many Raters.” PsychologicalBulletin. 76, 378–382.

Fleiss, J.L., Cohen, J., Everitt, B.S. (1969). “Large-Sample Standard Errors of Kappa and WeightedKappa.” Psychological Bulletin. 72, 323–327.

Fleiss, J.L., Cohen, J. (1973). “The Equivalence of Weighted Kappa and the Intraclass CorrelationCoefficient as a Measure of Reliability.” Educational and Psychological Measurement. 33,613–619.

Fleiss, J.L. (1981). Statistical Methods for Rates and Proportions. Second Edition. New York: Wiley.Graham, P., Bull, B. (1998). “Approximate Standard Errors and Confidence Intervals for Indices of

Positive and Negative Agreement.” Journal of Clinical Epidemiology. 51, 763–771.

Page 397: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

384 Pharmaceutical Statistics Using SAS: A Practical Guide

Grove, W.M., Andreasen, N.C., McDonald-Scott, P., Keller, M.B., Shapiro, R.W. (1981).“Reliability Studies of Psychiatric Diagnosis: Theory and Practice.” Archives of GeneralPsychiatry. 38, 408–413.

Guyatt, G., Patrick, D., Feeny, D. (1991). “Glossary.” Controlled Clinical Trials. 12, 274S–280S.Hamilton, M. (1960). “A Rating Scale for Depression.” Journal of Neurology, Neurosurgery and

Psychiatry. 23, 56–62.Hatcher, L. (1994). A Step-by-Step Approach to Using the SAS System for Factor Analysis and

Structural Equation Modeling. Cary, NC: SAS Institute Inc.International Conference on Harmonization (ICH). (1998). “Statistical Principles for Clinical

Trials.” E-9 Document. Available at http://www.ich.org/LOB/media/MEDIA485.pdf.Johnson, R.A., Wichern, D.W. (1982). Applied Multivariate Statistical Analysis. Englewood Cliffs,

NJ: Prentice-Hall.Kraemer, H. (1991). “To Increase Power in Randomized Clinical Trials without Increasing Sample

Size.” Psychopharmacology Bulleton. 27, 217–224.Kratochvil, C.J., Perwien, A., Vaughan, B., Faries, D., Saylor, K., Busner, J., Buermeyer, C.,

Hin-Hong, W., Swindle, R. (2004). “Emotional Expression and ADHD Pharmacotherapy inChildren: A New Measure.” American Academy of Child and Adolescent Psychiatry. AACAPposter presentation.

Landis, J.R., Koch, G.G. (1977). “The Measurement of Observer Agreement for Categorical Data.”Biometrics. 33, 159–174.

Leon, A.C., Marzuk, P.M., Portera, L. (1995). “More Reliable Outcome Measures Can ReduceSample Size Requirements.” Archives of General Psychiatry. 52, 867–871.

Lin, L.I. (1989). “A Concordance Correlation Coefficient to Evaluate Reproducibility.” Biometrics.45, 255–268.

Lin, L., Hedayat, A.S., Sinha, B., Yang, M. (2002). “Statistical Methods in Assessing Agreement:Models, Issues, and Tools.” Journal of the American Statistical Association. 97, 257–270.

Mehta, C.R., Patel, N.R. (1983). “A Network Algorithm for Performing Fisher’s Exact Test in r × cContingency Tables.” Journal of the American Statistical Association. 78, 427–434.

Patrick, D.L., Martin, M.L., Bushnell, D.M., Yalcin, I., Wagner, T.H., Buesching, D.P. (1999).“Quality of life for women with urinary incontinence: further development of the incontinencequality of life instrument.” Urology. 53, 71–76.

Perrin, E.S., Aaronson, N.K., Alonso, J., Burnam, A., Lohr, K., Patrick, D.L. (1997). InstrumentReview Criteria. Medical Outcomes Trust, March.

Sanchez, M.M., Binkowitz, B.S. (1999). “Guidelines for Measurement Validation in Clinical TrialDesign.” Journal of Biopharmaceutical Statistics. 9, 417–438.

Sandifer, M.G., Hordern, A.M. Timbury, G.C., Green, L.M. (1968). “Psychiatric diagnosis: Acomparative study in North Carolina, London, and Glasgow.” British Journal of Psychiatry.114, 1–9.

Shrout, P.E. (1998). “Measurement reliability and agreement in psychiatry.” Statistical Methods inMedical Research. 7, 301–317.

Shrout, P.E., Fleiss, J.L. (1979). “Intraclass correlations: Uses in assessing rater reliability.”Psychological Bulletin. 86, 420–428.

Spitznagel, E.L., Helzer, J.E. (1985). “A Proposed Solution to the Base Rate Problem in the KappaStatistic.” Archives of General Psychiatry. 42, 725–728.

Stokes, M.E., Davis, C.S., Koch, G.G. (2000). Categorical Data Analysis Using the SAS System,Second Edition. Cary, NC: SAS Institute Inc.

Stratford, P.W., Binkley, J.M., Riddle, D.L. (1996). “Health Status Measures: Strategies andAnalytic Methods for Assessing Change Scores.” Physical Therapy. 76, 1109–1123.

Thurber, S., Snow M., Honts, C.R. (2002). “The Zung Self-Rating Depression Scale—ConvergentValidity and Diagnostic Discrimination.” Assessment. 9, 401–405.

Wagner, T.H., Patrick, D.L., Bavendam, T.G., Martin, M.L., Buesching, D.P. (1996). “Quality oflife of persons with urinary incontinence: development of a new measure.” Urology. 47, 67–72.

Page 398: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14

Decision Analysis in DrugDevelopment

Carl-Fredrik BurmanAndy GrieveStephen Senn

14.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 385

14.2 Introductory Example: Stop or Go?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .386

14.3 The Structure of a Decision Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 392

14.4 The Go/No Go Problem Revisited . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394

14.5 Optimal Sample Size . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 397

14.6 Sequential Designs in Clinical Trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 406

14.7 Selection of an Optimal Dose . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412

14.8 Project Prioritization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421

14.9 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 426

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 426

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 426

Decision analysis is a quantitative methodology for analyzing and optimizing decisions. Ithas its roots in statistical decision theory as outlined by Raiffa and Schlaifer (1961). Thebasic idea is to structure the decision problem, quantify uncertainties and preferences, andthen solve the resulting optimality problem. In this chapter, we will present some typicaldecision problems in drug development: go/no go decisions, sample size calculations,dose-finding studies, sequential designs, and project prioritization. The examples will bedisplayed and analyzed using the dedicated Operations Research module SAS/OR, togetherwith optimization routines in SAS/IML, as well as the statistician’s standard SAS arsenal.

14.1 IntroductionWe all make thousands of decisions every day. Decision-making is part of most areas of life.Consequently, decision analysis has applications in many areas of society, business, and,not least, medicine. Within the medical fields, decision analysis has, quite naturally, mostoften been used in order to find the optimal strategy for diagnosing and treating patients

Carl-Fredrik Burman is Statistical Science Director, Technical and Scientific Development, AstraZeneca, Sweden. AndyGrieve is Head, Statistical Research and Consulting Center, Pfizer, United Kingdom. Stephen Senn is Professor, Departmentof Statistics, University of Glasgow, United Kingdom.

Page 399: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

386 Pharmaceutical Statistics Using SAS: A Practical Guide

(e.g., Parmigiani, 2002, Chapters 5–6; Bertolli et al., 2003; Liu et al., 2003). The analysis isoften made for a population although attempts have been made to also include thepreferences of the individual patient (Protheroe et al., 2000; Elwyn et al., 2001). In healtheconomics, the benefits of a treatment are weighed against its costs, and these componentsare often embedded in a decision analysis model (Cooper et al., 2002; Brown et al., 2003).

Whereas there are plenty of decision analytic applications to medical decision-makingand cost-effectiveness analysis in the literature, less has been written about suchapplications to the core drug development process. One reason is probably theconfidentiality of internal company decisions. However, as identified in the FDA’s recentCritical Path Initiative (FDA, 2004, 2006), drug development faces large and growingproblems, including increased development costs and fewer new drugs that reach themarket. One part of the solution to the industry’s productivity problem, we believe, isthrough model-based approaches (FDA, 2004, p. 24) coupled with a decision analysis(Poland and Wada, 2001; Burman et al., 2005). This chapter will give a few examples ofdecision analysis applications from different parts of drug development, often promotingbut sometimes warning against their use. Special attention will be given to the use ofdecision analysis during clinical development since the use of clinical trials is not only oneof the cornerstones of drug development, it is also something that adds a special structureto the problems and thereby differentiates the decision problems in clinical developmentfrom those in other parts of science and business.

It is important to recognize that there are many stakeholders in the process ofdeveloping a drug and distributing it to the patients in need. It is interesting to comparewhat is optimal for different stakeholders. Of course, we ought to have a system whereindustry investments, regulations, evaluations by health care providers, patent laws,guidelines, prescription habits, etc., all serve the good of the patients. The decisions ofdifferent agents obviously interact with each other. This chapter will mainly have anindustry perspective. However, now and then we will touch upon some other perspectivesand the interaction between the interests of different stakeholders.

14.1.1 OutlineWe will begin with an introductory example considering a very simplified clinical program(Section 14.2). This example will introduce some of the ideas and notions. In Section 14.3,we will describe the fundamentals of classical statistical decision theory. We will then beready to describe a number of different applications of decision analysis in drugdevelopment:

• Go/no go problems (Section 14.4).• Sample size calculation from a perspective of maximizing company profits (Section 14.5).• Sequential design of clinical trials (Section 14.6).• Finding the best dose in a dose-response study (Section 14.7).• Project prioritization (Section 14.8).

14.2 Introductory Example: Stop or Go?Decision trees are very useful for analyzing decision problems. The DTREE procedure canbe used to construct, plot, and evaluate such decision trees. In some cases there is anenormous number, or even a continuum, of decision alternatives available. Such problemscan often be attacked with optimization routines and/or simulations (see Sections 14.5and 14.7). In this section, we will introduce a very simple decision tree example, which willbe expanded in Sections 14.4 and 14.5.

A classic decision problem is whether to invest or not. In a drug development setting,the question might be if a large and expensive Phase III program should be launched or if

Page 400: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 387

the development of the drug should be discontinued. The consequences of a discontinuationare fairly easy to assess. There is, however, considerable uncertainty in the results of aPhase III program, comparing a new drug candidate with the current standard therapy. Tosimplify things, the result might be that the new drug is regarded to be either superior,non-inferior (but not superior), or inferior to the active control. The new drug cannot bemarketed in case of inferiority, as regulatory approval will then not be given. Superior ornon-inferior efficacy results will both lead to marketing. However, the regulatory labelingwill be different and the market appreciation and sales are likely to be widely different.

Program 14.1 uses PROC DTREE in SAS/OR to create a decision tree for thedescribed go/no go problem. The STAGE1 data set describes the structure of the tree.Each of the five rows in this data set corresponds to a branch, and the four variables definetree characterisics such as node types and branch labels. For example, the STTYPEvariable assumes two values, D and C, that identify the decision and chance nodes,respectively. The decision node is a decision to conduct the study or not and the chancenode is a (random) outcome of the trial (superiority, non-inferiority, or inferiority). TheOUTCOME variable specifies labels for the five branches in this decision tree.

Program 14.1 Simple go/no go problem with an efficacy outcome variable

data stage1;length _outcome_ $6.;input _stname_ $ _sttype_ $ _outcome_ $ _success_ $;datalines;Decision D No_go .. . Go DevelopDevelop C Super .. . Noninf .. . Infer .;

* Trial’s outcome;symbol1 value=triangle height=10 color=black width=3 line=1;* Decision point;symbol2 value=square height=10 color=black width=3 line=1;* End nodes;symbol3 value=none height=10 color=black width=3 line=1;proc dtree stagein=stage1;

treeplot/graphics norc nolegendlinka=1 linkb=2 symbold=2 symbolc=1 symbole=3;run;quit;

The decision tree generated by Program 14.1 is displayed in Figure 14.1. This decisiontree is almost the simplest one that is meaningful. In general, decision trees that describethe clinical development of a drug involve multiple decisions and have a lot more decisionand/or chance nodes. We will consider decision trees arising in clinical trials further inSection 14.4. Here we will refine the tree somewhat on the consequence side.

Efficacy of a drug is not everything. Safety is also important. Assume, for example, thatthe competitor drug is connected to a specific type of adverse events (AE). Our new drugmay or may not have the advantage of a considerably lower rate of these AEs, dependingon whether the same biological pathway leading to the safety problem is triggered. We willignore the possibility of getting a higher rate of the AE, as it is assumed that the safetyproblem is zero-one. It is, however, straightforward to include a possibility of a safetydisadvantage in the analysis.

Page 401: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

388 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 14.1 Decision tree in the simple go/no go problem. The square node is the decision to conduct the trialor not and the triangle node is the trial’s efficacy outcome (superiority, non-inferiority, or inferiority).

No_go

Go Super NoninfInfer

Program 14.2 produces a decision tree (Figure 14.2) which also reflects the outcome ofthe adverse event comparison in the Phase III trial. The STAGE2 data set defines sevenbranches and three nodes:

• DECISION (decision node): A decision to conduct the trial or not.• DEVELOP (chance node): Outcome of the efficacy analysis (superiority, non-inferiority,

or inferiority).• AE (chance node): Outcome of the safety analysis (superiority or equivalence).

The tree does not consider the AE comparison when the experimental drug is inferior tothe competitor drug in terms of efficacy because the inferiority outcome is assumed toimply that regulatory approval will not be achieved.

Program 14.2 Simple go/no go problem with multiple outcome variables (efficacy and safety)

data stage2;length _outcome_ $15.;input _stname_ $ _sttype_ $ _outcome_ $ _success_ $;datalines;Decision D No_go .. . Go DevelopDevelop C Eff_super AE. . Eff_noninf AE. . Eff_infer .AE C AE_super .. . AE_equal .;

* Trial’s outcome;symbol1 value=triangle height=10 color=black width=3 line=1;* Decision point;symbol2 value=square height=10 color=black width=3 line=1;* End nodes;symbol3 value=none height=10 color=black width=3 line=1;proc dtree stagein=stage2;

treeplot/graphics norc nolegendlinka=1 linkb=2 symbold=2 symbolc=1 symbole=3;run;quit;

Page 402: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 389

Figure 14.2 Decision tree in the simple go/no go problem with two outcome variables. The square node is thedecision to conduct the trial or not and the triangle nodes represent the trial’s efficacy outcome (superiority,non-inferiority, or inferiority) and safety outcome (superiority or equivalence).

No_go

Go

Eff_super

Eff_noninf

Eff_infer

AE_super AE_equal AE_super AE_equal

14.2.1 Evaluating a Decision TreeStructuring a problem is an essential part in solving it. In the above example, this step wasrather trivial. For a complex problem, however, it can be quite difficult to find a gooddescription which is as simple as possible and yet captures the essence of the problem.Given the tree structure of a problem, the optimal decision will sometimes be evident.However, it is often necessary to assess the consequences and uncertainties quantitatively.

In the Phase III investment decision, let us say that we can estimate the probabilities ofsuperiority, non-inferiority, and inferiority outcomes in a Phase III trial. This estimation ofprobabilities is, of course, related to power calculations and depends on a number offactors, such as the precise definition of non-inferiority, the assumed effect size, andvariability, etc. We will come back to such topics later and for now take the computation ofthese probabilities for granted. Uncertainties in a regulatory response and sales can also beincluded in a decision analysis model but we will ignore these aspects for now. We will alsoignore any other potential safety issues apart from the specific kind of AE underconsideration.

Let E+, E=, and E− denote the events of superiority, non-inferiority, and inferiority,respectively. Similarly, denote the event of proven safety benefit of the new drug withrespect to the specified AE by S+ and the complement event (no proven safetyimprovement) by S=. For the following example, let

P (E+) = 0.1, P (E=) = 0.4, P (E−) = 0.5.

We assume that the outcome for the specified AE is independent of the effect outcome andthat

P (S+) = 0.7, P (S=) = 0.3.

The independence may not, of course, be the case in practice and the joint probability ofefficacy and safety may have to be considered instead. (Compare the conditioning in asimilar situation in Program 14.4.)

Note that the probabilities given are for the different conclusions from the Phase IIIprogram. It is quite possible that superiority cannot be concluded even if the new drug infact produces a better effect than the competitor. The probabilities are included in thePROB3 data set in Program 14.3.

In order to compare the alternatives we also need to quantify the value of the possibleoutcomes. Immediate discontinuation of the project is likely to mean ignorable costs andincomes from the drug (we will not consider the possibilities of out-licensing or limited

Page 403: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

390 Pharmaceutical Statistics Using SAS: A Practical Guide

development). We will therefore regard the discontinuation possibility to have value 0.Previous costs or future unavoidable costs are of no interest in the decision problem. Whatmatters is, of course, only the costs and gains which can be influenced by the decisionsmade now or in the future.

Expected net profit is taken as the optimality criterion. A more sophisticated analysisshould consider some additional factors:

• A small expected gain from an investment may not be worth the risk of losing theinvestment. The degree of risk aversion is likely to be larger in smaller companies, forwhich a failure in Phase III may jeopardize the company’s survival.

• The resources (e.g., money or personnel) may be limited, forcing the company to choosebetween several promising projects (see Section 14.8).

• Future cash-flows should be discounted by an appropriate rate. The discount rate shouldprimarily reflect the cost of capital, e.g., the interest rate paid. Some organizationsassume a higher discount rate in order to also account for the investment risk and/orlimited personnel resources. Although such high discount rates might be used as asimplifying tool, we do not recommend them for important decisions. It is better toexplicitly model resource restrictions and risk aversion.

One way of dealing with risk is to define the utility as an appropriate concave functionof money (Raiffa, 1968, Chapter 4) and use the expected utility as the optimality criterion.For the sake of simplicity, however, we will assume that cash-flows are discounted and thatrisk aversion and resource restrictions can be ignored.

Suppose that the cost of running the Phase III program is 250 (the monetary unit inthis example can be a million of US dollars, or MUSD). An inferior drug will not bepossible to sell and therefore produces no income. Non-inferiority is enough to get somesales but the income will be considerably higher in the case of superiority. Ignoring thedevelopment costs, we assume the net profit, as a function of the Phase III programoutcome, to be G(·). The net profit here denotes the sales minus promotional andmanufacturing costs over the drug’s life-cycle. We will let

G(E+, S+) = 1200, G(E+, S=) = 550, G(E=, S+) = 450, G(E=, S=) = 100

and, as stated earlier, G(E−, ·) = 0.Program 14.3 creates a decision tree for the introduced decision analysis problem. The

development costs are specified in the STAGE3 data set and the gains are included in thePAYOFF3 data set.

Program 14.3 Evaluated decision tree in the simple go/no go problem with multiple outcome variables(efficacy and safety)

data stage3;length _outcome_ $10.;input _stname_ $ _sttype_ $ _outcome_ $ _reward_ _success_ $;datalines;Phase3 D No_go . .. . Go -250 DevelopDevelop C Eff_super . AE. . Eff_noninf . AE. . Eff_inf . .AE C AE_super . .. . AE_equal . .;

Page 404: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 391

data prob3;length _event1_ _event2_ _event3_ $10.;input _event1_ $ _prob1_ _event2_ $ _prob2_ _event3_ $ _prob3_ ;datalines;Eff_super 0.2 Eff_noninf 0.5 Eff_inf 0.3AE_super 0.3 AE_equal 0.7 . .;

data payoff3;length _state1_ _state2_ $10.;input _state1_ $ _state2_ $ _value_;datalines;Eff_super AE_super 1200Eff_super AE_equal 550Eff_noninf AE_super 450Eff_noninf AE_equal 100Eff_inf . 0;

* Trial’s outcome;symbol1 value=triangle height=10 color=black width=3 line=1;* Decision point;symbol2 value=square height=10 color=black width=3 line=1;* End nodes;symbol3 value=none height=10 color=black width=3 line=1;proc dtree stagein=stage3 probin=prob3 payoffs=payoff3;

ods select parameters policy;treeplot/graphics norc nolegend compresslinka=1 linkb=2 symbold=2 symbolc=1 symbole=3;evaluate/summary;run;quit;

Output from Program 14.3

Decision Parameters

Decision Criterion: Maximize Expected Value (MAXEV)Optimal Decision Yields: 1.5

Optimal Decision Policy

Up to Stage Phase3

Alternatives Cumulative Evaluatingor Outcomes Reward Value----------------------------------------No_go 0 0.0Go -250 251.5*

Figure 14.3 displays the decision tree generated by Program 14.3, and cumulativerewards (CR) and expected values (EV) for each branch of the tree. Output 14.3 showsthat the optimal value for the go option is +1.5 MUSD. This value is the result of acumulative cost (negative reward) of 250 MUSD for running the trial and an evaluatedexpected value at the end node of 251.5 MUSD. The stop option has a zero value.

It should be noted that there are many possible descriptions of the same problem.Instead of using separate chance nodes for efficacy and safety outcomes, one can, of course,

Page 405: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

392 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 14.3 Evaluated decision tree in the simple go/no go problem with two outcome variables. The squarenode is the decision to conduct the trial or not and the triangle nodes represent the trial’s efficacy outcome(superiority, non-inferiority, or inferiority) and safety outcome (superiority or equivalence).

cr= 0 EV= 0

No_go

cr= 0 EV= 0

Go

cr=-250 EV= 0

Eff_super

cr=-250 EV= 0

Eff_noninf

cr=-250 EV= 0

Eff_inf

cr=-250 EV= 0

AE_super

cr=-250 EV= 1200 AE_equal

cr=-250 EV= 550 AE_super

cr=-250 EV= 450 AE_equal

cr=-250 EV= 100

consider only one chance node with an outcome space consisting of five combined events:{E+, S+}, {E+, S=}, {E=, S+}, {E=, S=} and {E−}. Different levels of refinement arepossible, and more or less suitable according to the situation. For example, one can averageover all possible safety outcomes and just use the probabilities for the efficacy outcomestogether with the expected gains given these outcomes, i.e.,

G(E+) = G(E+, S+)P (S+) + G(E+, S=)P (S=)

and

G(E=) = G(E=, S+)P (S+) + G(E=, S=)P (S=).

This problem formulation would be adequate at the moment. However, the simple decisionproblem that we have analyzed in this section will be expanded in Section 14.4 and theseparation between efficacy and safety outcomes will then be crucial. The previousassumption about independence of efficacy and safety results was made for convenience.Dependence is easily treated by providing conditional probabilities in the data set specifiedin the PROBIN option of PROC DTREE. This is shown in Program 14.4 (see Section 14.4)in another situation of dependence.

Before expanding the drug development problem, we will give a short generaldescription of decision analysis.

14.3 The Structure of a Decision AnalysisThe foundations of statistical decision theory were laid by Wald (1950) and Savage (1954).The theory was mainly concerned with problems within statistical inference, such asestimation and hypothesis testing, rather than with real-life decision problems. A standardformulation (Berger, 1985; Lehmann, 1986; French and Rıos Insua, 2000) is that θ denotesthe unknown parameter (or “state of nature”). After observing a random vector X, whichdistribution is determined by θ, the decision-maker is to make a decision (take an action)denoted by d(X). Statistical decisions may be related to accepting or rejecting a hypothesisor to choosing a point estimate of θ. The consequences of different decisions are modeled by

Page 406: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 393

a fully specified loss function L(θ, d(X)). One may proceed by calculating the risk function

r(θ, d) = EX [L(θ, d(X))],

where the expectation is taken over the distribution of X for the parameter θ.Since the risk function depends on the unknown parameter, it is not clear which decision

rule is optimal. Two possible criteria for optimality are the minimax and Bayes criteriadefined below.

Minimax criterion. The minimax rule chooses the decision d = d(X) that minimizesmaxθ r(θ, d).

Bayes criterion. The Bayes solution is based on a prior distribution π(θ) for θ andchooses the decision rule that minimizes

Eθ[r(θ, d)] =∫

r(θ, d)π(θ) dθ.

Statistical decision theory can also be applied to decision problems which we do notnecessarily think of as “statistical”, such as deciding whether to invest or not (Raiffa,1961). In these problems the temporal order is often different from the typical statisticalproblem: it is common that a decision d (e.g., a go/no go decision in a Phase III trial) hasto be taken before observing a random outcome X (e.g., trial results). The decision-maker’spreferences are often modeled as a utility u = u(θ, d,X). To model a utility u instead of aloss L is purely conventional as they are easily interchangeable by the relation

u = constant − L.

It is also common to see sequential decision problems. There are often several decisionsd1, d2, . . . which are to be taken. Between the decision points, more and more observationsX1, X2, . . . can be collected. Each Xk may be a random vector whose distribution isdetermined by θ. The random outcome Xk can also depend on earlier decisions d1, . . . , dk−1,as these may relate to which experiments are run. On the other hand, the decisions dependon previously collected information, dk+1 = dk+1(X1, . . . , Xk). This structure can, at leastfor simple discrete problems, be illustrated as a decision tree with a number of decisionnodes as well as chance nodes.

By writing d = {d1, d2, . . .} and X = {X1, X2, . . .}, we still have a utility function of theform u = u(θ, d,X). Given a prior distribution for θ, say, π(θ), and using expected utility asthe optimality criterion, the problem is to find the decision strategy d which maximizes

E[u(d, X)] =∫

u(θ, d,X)π(θ) dθ.

Backward induction is often useful to solve this problem (Bather, 2000). The minimaxoptimality criterion, although sometimes fruitful for some statistical problems, is usually oflimited value in a real-life decision problem. For example, the minimax solution of theinvestment problem is almost invariably not to invest. This philosophy could be parodiedas one which leads one to spend one’s life in bed for fear of being run over by a car if oneleaves the house.

A main criticism of fully Bayesian methods that rely on informative priors has been thesubjective component. Results that strongly depend on the experimenter’s prior beliefs arelikely not to be fully accepted by other stakeholders or by the scientific community. Thiscriticism, however, loses its strength when the conclusions are not meant to convinceindividuals other than those who make the assumptions. Therefore, a semi-Bayesianapproach is reasonable: use Bayesian methods internally but classical frequentismexternally. For example, a company might use its prior opinion of the effect of a new drug

Page 407: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

394 Pharmaceutical Statistics Using SAS: A Practical Guide

in order to decide on how much resources, if any, to put into its development, how clinicaltrials should be designed and dimensioned, etc. Still, the company may give traditionalfrequentist statistical analyses to the regulatory bodies and to the scientific communitywhen publishing the results. As we will see, the utility may even be constructed as afunction of the result of the frequentist analysis.

The term “Decision Analysis” was coined in 1965 by Howard (Howard and Matheson,1984, pages 3 and 97). Decision analysis is partly the application of statistical decisiontheory but does also pay attention to structuring and modeling a problem. These issues aretypically more complicated than the computational optimization, given a specified model.Good accounts of decision analysis are provided by Raiffa (1968) and in a collection ofpapers edited by Howard and Matheson (1984).

14.4 The Go/No Go Problem RevisitedThe go/no go example of Section 14.2, having only two available alternatives, is simplistic.There is usually a larger set of decision alternatives. The company may try to out-licensethe drug. It can decide to postpone the decision so that more information regarding thedrug’s likely effect and market potential can be collected. Even if the decision is to start aPhase III program immediately, there are different possible designs for the program.Important design factors are which dose(s) to use and the sample size. All these examplesmay be modeled by including more decision nodes in the decision tree. In this section wewill illustrate the possibility of adding decision nodes with a problem involving the optionto purchase information regarding an important AE.

Recall from Section 14.2 that there is good hope that the new drug in the example hasan advantage over the existing therapy with respect to a certain adverse event. Assumethat a preliminary test (pretest) can be run to investigate this further. If the pretest ispositive, then it is highly likely that an AE advantage can be demonstrated in Phase IIIdevelopment. A negative result, on the other hand, predicts no advantage. The question iswhether it is worth a relatively modest cost to perform the pretest? We will assume thatthis investigation may be run in parallel with other necessary activities before making thedecision on whether to proceed to a Phase III program (if this were not the case, andrunning the pretest would delay the project, the payoff data set should reflect that thereward depends on the time of marketing).

One can think of a number of different investigations that could, depending on thesituation, possibly serve as pretests, e.g.:

• Use of an AE animal model.• Studies of the binding between drug and receptor.• A clinical trial of limited size and duration, perhaps focusing on a surrogate marker in a

selected high-risk population.

Often, the results of such investigations are not dichotomous but may be more or lesspositive or negative. For simplicity, however, we assume that only two outcomes(positive/negative) are possible.

Program 14.4 analyzes the same problem as in Program 14.3 but with the addition of thepretest option. It is assumed that the cost of the pretest is 20 MUSD and that a positive ornegative pretest predicts that superiority with respect to the AE can be shown in Phase IIIwith probabilities 0.9 and 0.15, respectively. In order to be consistent with the problemconsidered in Program 14.3, the probability for a positive pretest must be 0.2, since

P (S+) = P (S+ | Positive pretest) P (Positive pretest)

+ P (S+ | Negative pretest) P (Negative pretest).

Page 408: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 395

Program 14.4 Evaluated decision tree in the simple go/no go problem with two outcome variables (efficacyand safety) and a pretest

data stage4;length _stname_ _outcome_ _success_ $10.;input _stname_ $ _sttype_ $ _outcome_ $ _reward_ _success_ $;datalines;Pretest D No_test 0 Phase3. . Test -20 AEtestAEtest C AEpos . Phase3. . AEneg . Phase3Phase3 D No_go 0 .. . Go -250 DevelopDevelop C Eff_super . AE. . Eff_noninf . AE. . Eff_inf . .AE C AE_super . .. . AE_equal . .;

data prob4;length _given1_ _given2_ _event1_ _event2_ _event3_ $10.;input _given1_ $ _given2_ $ _event1_ $ _prob1_

_event2_ $ _prob2_ _event3_ $ _prob3_;datalines;. . AEpos 0.2 AEneg 0.8 . .. . Eff_super 0.2 Eff_noninf 0.5 Eff_inf 0.3No_test . AE_super 0.30 AE_equal 0.70 . .Test AEpos AE_super 0.90 AE_equal 0.10 . .Test AEneg AE_super 0.15 AE_equal 0.85 . .;

data payoff4;length _state1_ _state2_ $10.;input _state1_ $ _state2_ $ _value_;datalines;Eff_super AE_super 1200Eff_super AE_equal 550Eff_noninf AE_super 450Eff_noninf AE_equal 100Eff_inf . 0;

* Trial’s outcome;symbol1 value=triangle height=10 color=black width=3 line=1;* Decision point;symbol2 value=square height=10 color=black width=3 line=1;* End nodes;symbol3 value=none height=10 color=black width=3 line=1;proc dtree stagein=stage4 probin=prob4 payoffs=payoff4;

ods select parameters policy;treeplot/graphics norc nolegend compresslinka=1 linkb=2 symbold=2 symbolc=1 symbole=3 display=(link);evaluate/summary;run;quit;

Output 14.4 shows that the optimal decision path is to run the pretest and then go intoPhase III development only if the pretest is positive. Comparing the optimal values inOutput 14.3 and Output 14.4, we see running the pretest increases the expected value of

Page 409: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

396 Pharmaceutical Statistics Using SAS: A Practical Guide

the project from 1.5 MUSD to 16.9 MUSD. The decision tree produced by Program 14.4 isnot shown in order to save space.

Output from Program 14.4

Decision Parameters

Decision Criterion: Maximize Expected Value (MAXEV)Optimal Decision Yields: 16.9

Optimal Decision Policy

Up to Stage Pretest

Alternatives Cumulative Evaluatingor Outcomes Reward Value----------------------------------------No_test 0 1.5Test -20 36.9*

Optimal Decision Policy

Up to Stage Phase3

Cumulative EvaluatingAlternatives or Outcomes Reward Value

------------------------------------------------------------No_test No_go 0 0.0No_test Go -250 251.5*Test AEpos No_go -20 0.0Test AEpos Go -270 434.5*Test AEneg No_go -20 0.0*Test AEneg Go -270 205.8

It is often crucial to investigate the robustness of the conclusions. The modelparameters, especially rewards and probabilities, are typically uncertain. It is good practiceto vary the most important parameters and look at how these variations affect the analysis.PROC DTREE is an interactive procedure and allows the user to modify the rewards withthe MODIFY statement (see Program 14.17).

However, the most powerful way of investigating robustness properties is via macroprogramming. A simple SAS macro that evaluates the robustness of the decision analysismodel is given in Program 14.5. In this macro, all payoffs are scaled by a common factor f .The output of the program (not displayed) shows that the optimal decision patternchanges considerably when the payoff factor f is varied:

• For sufficiently small payoffs, say, f = 0.5, the expected value of a Phase III trial isnegative even if the drug has an AE advantage.

• When f = 0.8, the AE advantage would motivate a Phase III trial. However, the cost ofthe pretest is too high in comparison with the information it will provide.

• In the standard scenario, f = 1.0, it is optimal to run the pretest and run a Phase IIItrial if and only if the pretest result is positive.

• For f = 1.2, the pretest still gives valuable information but the optimal strategy is to goto Phase III development directly, avoiding the pretest cost.

Page 410: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 397

• Finally, for really high payoffs (e.g., f = 1.5), it is worthwhile to run a Phase III trialeven when the pretest is negative. Since the Phase III trial will be done irrespective ofthe result of the pretest, the pretest is obviously redundant in this model and with theseparameters. Thus, the analysis shows that the optimal decision is to conduct a Phase IIItrial without a previous pretest. Note, however, that a pretest may help optimize thedesign of the Phase III trial. If this is the case, an extended model may still show thatthe pretest has value.

Program 14.5 Robustness check when varying payoffs

/* Refers to data sets from the previous program */%macro robust(payoff_factor);

data payoff_changed;set payoff4;_value_=&payoff_factor*_value_;

proc dtree stagein=stage4 probin=prob4 payoffs=payoff_changed criterion=maxev;evaluate/summary;run;quit;

%mend robust;

%robust(0.5);%robust(0.8);%robust(1.0);%robust(1.2);%robust(1.5);

14.5 Optimal Sample SizeThe Phase III investment example resembles investment problems from other industries,e.g., oil drilling examples described in Raiffa and Schlaifer (1961, Section 1.4.3). Oneimportant difference is that Phase III development consists of experiments which we are todesign and whose results can be modeled with standard statistical methods. The power, orprobability of showing superiority, in one clinical trial is easily determined given theresponse distributions and test method. The response distributions are typicallydetermined by parameters such as mean effect and standard deviation. Furthermore, theseparameters can be modeled using previous clinical (and sometimes preclinical) data (Pallayand Berry, 1999; Burman et al., 2005).

So far, in Sections 14.2 and 14.4, we have taken the Phase III program to be fixed (if itis run at all). The design of these trials is, however, of utmost importance and definitely afield where statisticians have a lot to contribute. In this section we will consider one singledesign feature: the trial’s sample size.

Decision analytic approaches to sample size calculations in clinical trials have beenconsidered in a variety of situations. General Bayesian approaches are outlined by Raiffaand Schlaifer (1961, Section 5.6) and Lindley (1997). Claxton and Posnett (1996) as well asO’Hagan and Stevens (2001) apply such ideas to clinical trials with health economicsobjectives. Gittins and Pezeshk (2000) and Pallay (2000) discuss the sizing of Phase IIItrials, considering regulatory requirements. A review of Bayesian approaches to clinicaltrial sizing is provided by Pezeshk (2003).

Sample size has often been viewed as a relatively simple function of the followingparameters:

• “Least clinically relevant effect” δ.• Standard deviation σ.

Page 411: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

398 Pharmaceutical Statistics Using SAS: A Practical Guide

• Power 1 − β.• One-sided significance level α/2.

In a two-arm trial of reasonable size, so that a normal approximation is appropriate, thetotal sample size is given by

n =4 (Φ−1(1 − α/2) + Φ−1(1 − β))2

(δ/σ)2 ,

where Φ(·) is the standard normal cumulative distribution function.The main difficulty in sample size calculations is the choice of α, β, δ, and σ. The

one-sided significance level α is conventionally set at a 2.5% level so that the two-sided sizeof the test is 5% (FDA, 1998). The standard deviation σ is sometimes hard to estimate atthe planning stage and a misspecification may distort the resulting sample sizeconsiderably. However, there is often enough data from previous smaller trials with the testdrug and from Phase III trials of other drugs with the same response variable for areasonably robust guess of the value of σ.

More interesting parameters are β and, even more so, the treatment effect δ. We willstart by looking at the optimal power when δ is naıvely taken as a fixed known value. Afterthat we will discuss δ.

The power, 1 − β, is often conventionally set at 90%. This is indeed convenient for thestatistician performing the sample size calculation as he or she does not have to considerthe context around the trial. The importance of the trial, cost of experimentation,recruitment time and other factors are simply ignored. We argue, however, that the choiceof the sample size is often a critical business decision. A Phase III program or even a singlemortality trial will often cost in the order of 100 MUSD (DiMasi et al., 2003). Changingthe power from 80% to 90%, or from 90% to 95%, can result in a considerable increase ofthe trial’s cost, sometimes tens of MUSD. In addition, and often even more important, thetime to marketing may be delayed. For such important decisions, a decision analysisframework seems natural.

14.5.1 Optimal Sample Size Given a Fixed EffectWe will start with a very simple decision analysis model and then gradually add somefeatures that can increase the realism of the model. Suppose that a statistically significanttrial result in favor of the test drug will lead to regulatory approval and a subsequentmonetary gain G for the company. Think of G as the expected discounted net profit duringthe whole product life-cycle, where sale costs (production, distribution, promotion, etc.) aresubtracted from the sales. In the case of a non-significant result, the drug will not beapproved and there will be no gain.

Assume that the cost for conducting a clinical trial with N > 0 patients isC(N) = C0 + cN , where C0 is the start-up cost and c is the cost per patient. If no trial isconducted, we have N = 0 and C(0) = 0. The trial cost and other development costs shouldnot be included in G. The company profit is assumed to be GI{S} − C(N), where I{S} = 1 ifthe trial is statistically significant and 0 otherwise. Note that this assumption is typicallynot realistic, as the magnitude of the estimated effect as well as the precision of theestimate are likely to affect the sales. Furthermore, the times of submission, approval, andmarketing are likely to depend on N . The model can be made more realistic byincorporating such aspects. This is partly done in Section 14.5.3 below.

Under these assumptions, the expected profit is given by

V (N) = E[GI{S} − C(N)] = Gp(N) − C(N),

where p(N) is the probability of a significant outcome, i.e., power. The values of theparameters vary considerably depending on the drug and therapeutic area. For the sake of

Page 412: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 399

illustration, assume that the effect size δ/σ = 0.2, value of the trial G = 1000, start-up costC0 = 50 and cost per patient c = 0.1. Program 14.6 computes the profit function for theselected parameter values.

Program 14.6 Computation of costs and gains in a Phase III trial

data npower;alpha=0.05; /* Two-sided significance level */critical=probit(1-alpha/2);effsize=0.2; /* Effect size */g=1000; /* Value of successful (i.e., significant) trial */startc=50; /* Start-up cost */c=0.1; /* Cost per patient */do n=0 to 2000 by 10;

power=probnorm(effsize*sqrt(n/4)-critical);egain=g*power;cost=startc+c*n;profit=egain-cost;output;

end;run;axis1 minor=none label=(angle=90 "Utility") order=(0 to 1000 by 200);axis2 minor=none label=("Sample size") order=(0 to 2000 by 500);symbol1 i=join width=3 line=1 color=black;symbol2 i=join width=3 line=20 color=black;symbol3 i=join width=3 line=34 color=black;proc gplot data=npower;

plot (cost egain profit)*n/overlay nolegend haxis=axis2 vaxis=axis1;run;quit;

Figure 14.4, generated by Program 14.6, depicts the resulting profit function. Themaximum profit is achieved at N = 1431 patients. This corresponds to the probability of asignificant outcome (power) of 96.6%.

Figure 14.4 Cost (solid curve), expected gain (dashed curve) and expected profit (dotted curve) as a functionof sample size

Util

ity

0

200

400

600

800

1000

Sample size

0 500 1000 1500 2000

It is clear that Program 14.6 relies on a fairly inefficient algorithm. It simply loopsthrough all values of N between 0 and 2000 to find the sample size that maximizes the

Page 413: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

400 Pharmaceutical Statistics Using SAS: A Practical Guide

expected net gain. A more efficient approach is to explicitly formulate this problem as anoptimization problem and utilize one of many optimization routines available in SAS. Inprinciple, it would have been possible to use a decision tree (i.e., PROC DTREE) for thisoptimization problem. However, since so many different values of N are possible, it is moreconvenient to treat it as an approximately continuous variable and use the optimizationprocedure PROC NLP.

The NOPT macro in Program 14.7 calculates the optimal sample size using the NLPprocedure. The procedure maximizes the value of PROFIT (expected net gain) withrespect to the N variable (total sample size). The underlying assumptions are identical tothe assumptions made in Program 14.6, i.e., the effect size δ/σ = 0.2, value of the trialG = 1000, start-up cost C0 = 50, and cost per patient c = 0.1.

Program 14.7 Optimal sample size

%macro nopt(effsize,g,startc,c,alpha);proc nlp;

ods select ParameterEstimates;max profit; /* Maximise the expected net gain */decvar n; /* Total sample size */critical=probit(1-&alpha/2);power=probnorm(&effsize*sqrt(n/4)-critical);egain=&g*power;cost=&startc+&c*n;profit=egain-cost;

run;%mend nopt;%nopt(effsize=0.2,g=1000,startc=50,c=0.1,alpha=0.05);

Output from Program 14.7

PROC NLP: Nonlinear Maximization

Optimization ResultsParameter Estimates

GradientObjective

N Parameter Estimate Function

1 n 1431.412256 1.336471E-9

Output 14.7 shows that the optimal sample size is 1431 patients. This value is identicalto the optimal sample size computed in Program 14.6. Some robustness checks are easilyimplemented by varying the assumed effect size. For example, it is easy to verify that theoptimal sample sizes for δ/σ = 0.15 and δ/σ = 0.25 are N = 2164 and N = 1018 patients,respectively.

The model used hitherto in this subsection is generally too simplistic to be of practicalvalue. We will return to the uncertainty in treatment effect in Section 14.5.3 but first, inthe next subsection, comment on the number of significant trials needed for regulatoryapproval.

14.5.2 The Optimal Number of TrialsOften two statistically significant trials are needed for regulatory approval. Assume thattwo virtually identical trials, each of size N , should be run and that both are required to besignificant.

Page 414: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 401

The reasoning used in Section 14.5.1 can then easily be extended to give

VTwo trials(N) = G · P (Both trials are significant) − 2C(N) = Gp(N)2 − 2(C0 + cN),

which can be optimized using a slightly modified version of Program 14.6.Provided that the two trials have equal conditions, it is optimal to have the same

sample size in both trials. Often the trials are, however, conducted in different countriesand in different populations. Costs, recruitment times, and anticipated effects maytherefore vary and non-equal sample sizes may be indicated.

The probability of achieving significant results in at least two trials is typically higher ifa fixed number of patients is divided into three trials instead of two. In fact, it is easier toget significances in two trials and supportive results in the third (defined as a pointestimate in favor of the new drug) when three trials are run than to get significances inboth trials when two trials are run. One numerical illustration is provided byProgram 14.8. Success of the trial program is defined as all trials giving positive pointestimates and at least two trials being statistically significant at the conventional level.

Program 14.8 Optimal number of trials

data no_of_trials;alpha=0.05;effect=20;sd=100;n_tot=2000;critical=probit(1-alpha/2);format m n 5.0 p_sign p_supp p_success 5.3;do m=1 to 5;

n=n_tot/m;p_sign=probnorm(effect/sd*sqrt(n/4)-critical);p_supp=probnorm(effect/sd*sqrt(n/4));p_cond=p_sign/p_supp;p_success=p_no_neg*(1-probbnml(p_cond,m,1));output;

end;label m=’Number of trials’

n=’Sample size’p_sign=’Prob of significance’p_supp=’Prob of supportive results’p_success=’Success probability’;

keep m n p_sign p_supp p_success;proc print data=no_of_trials noobs label;

run;

Output from Program 14.8

Number Prob ofof Sample Prob of supportive Success

trials size significance results probability

1 2000 0.994 1.000 0.0002 1000 0.885 0.999 0.7843 667 0.733 0.995 0.8164 500 0.609 0.987 0.7985 400 0.516 0.977 0.754

Output 14.8 lists several variables, including success probability when a total samplesize is divided equally in m trials (m = 1, . . . , 5). With a single trial, the probability of

Page 415: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

402 Pharmaceutical Statistics Using SAS: A Practical Guide

regulatory success is zero, as two significant trials are needed. Interestingly, the successprobability increases when going from two to three trials.

From a regulatory perspective, it is probably undesirable that sponsors divide a fixedsample size into more than two trials without any other reason than optimization of theprobability for approval. The regulators should therefore apply such rules that do notpromote this behavior. This is a simple example of the interaction between differentstakeholders’ decision analyses.

In fact, the reason that regulators require two trials to be significant is not entirelyclear. Say that two trials are run and each is required to be significant at a one-sided 0.025level (i.e., α = 1/40) and registration follows only when both trials are significant. Thiscorresponds to an overall Type I error rate of (1/40)2 = 1/1600 = 0.000625. However, ifcenters, patients, and protocols are regarded as exchangeable between the two trials, thenan equivalent protection in terms of Type I error rate can be provided to the regulatorusing fewer patients for equivalent overall power (Senn, 1997; Fisher, 1999; Rosenkranz,2002; Darken and Ho, 2004). In fact, one could run two such trials and simply pool theirresults together for analysis. The difference between the resulting pooled trial rule and theconventional two-trials rule in terms of the critical values of the standard normaldistribution is illustrated in the accompanying Figure 14.5 taken from Senn (1997).

In this figure, the boundaries for significance are plotted in the {Z1, Z2} space, where Z1is the standardized test statistic for the first trial and Z2 for the second. For the two-trialsrule we require that both Z1 > 1.96 and {Z2 > 1.96}. The resulting critical region is thatabove and to the right of the dashed lines. For the pooled trial rule we require(Z1 + Z2)/

√2 > 3.227. The latter rule may be derived by noting that Var(Z1 + Z2) = 2 and

1 − Φ(3.227) ≈ 0.000625. The associated critical region is above and to the right of the solidline.

Figure 14.5 Rejection regions for the pooled-trial and two-trials rules

z2

-2

0

2

4

6

z1

-2 0 2 4 6

If we calculate power functions for the pooled- and two-trials rules as a function of thestandardized non-centrality parameter, δ� for a single trial, then this has the value2δ�/

√2 =

√2δ� for the pooled-trial rule. Bearing in mind that both trials must be

significant for the two-trials rule, the power functions are

PowerTwo trials = (1 − Φ(1.96 − δ�))2

PowerPooled trial = 1 − Φ(3.227 −√

2 δ�).

Figure 14.6 displays a plot of the two power functions. It can be seen from the figurethat the power of the pooled-trial rule is always superior to that of the two-trials rule.

Page 416: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 403

Figure 14.6 Power of the pooled-trial (solid curve) and two-trials (dashed curve) rules

Pow

er

0.00

0.25

0.50

0.75

1.00

Treatment difference (delta)

0.0 0.5 1.0 1.5 2.0 2.5 3.0

One argument for preferring two trials to such a single trial would be if the trials werenot exchangeable and had different design details either in terms of type of centers andpatients or in terms of protocol. Two trials that were both significant at a 0.025 level wouldthen be more impressive in terms of robustness than a single trial significant at a 0.000625level. There is, however, no evidence that in their Phase III programs clinical trial sponsorsdeliberately choose trials that differ in design nor that regulators require them to. Forregulatory thinking on this issue, see Committee for Proprietary Medicinal Products(CPMP) (2001).

14.5.3 A Bayesian Approach to Sample Size CalculationsWe have, in Section 14.5.1, looked at the optimal sample size given a fixed treatment effectδ. However, the main problem in deciding on whether to conduct the trial at all and, inthat case, how to choose a sample size lies in the uncertainty regarding this parameter. If δcould be objectively established with good precision at this stage, there would be littleneed to conduct a clinical trial in order to estimate it. Hence, clearly we cannot insert thetrue value of δ in the power formula, if we regard this as being the effect of the treatment.It may also be misleading to insert a point estimate based on previous data, due to theuncertainty of this estimate. It is often recommended that the sample size calculationshould be based on the “least clinically significant effect” but even this is a quantity thatcannot be established with absolute precision, since physicians may disagree betweenthemselves and even from time to time, as to what it is.

To decide on whether to invest in a Phase III program a clinical trial sponsor mustclearly have some sort of prior opinion about either the size of the effect that would beimportant or the size that would be likely to obtain. We think that it is reasonable to tryto formalize this opinion using a Bayesian prior distribution. However, we warn here thatthe solution we describe below is not fully Bayesian and may be far from optimal becauseit does not model utility explicitly as a function of the treatment effect.

Fully Bayesian sample size determinations (with or without utility) have been discussedby several authors, e.g., Lindley (1997) and Pezeshk (2003). We will, however, investigatean approach where Bayesian decision theory is used only for in-house company decisions,such as designing the clinical program, see, for example, Gittins and Pezeshk (2000). Solelyfrequentist analyses of clinical trial results are communicated externally to regulators andthe public. This dichotomy between internal Bayesianism and external frequentism ismotivated by the stakeholder situation. The sponsor conducts the trial program in order to

Page 417: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

404 Pharmaceutical Statistics Using SAS: A Practical Guide

provide information to regulators and prescribing physicians on which they can base theirdecisions. These stakeholders will base their decisions on the data, for convenience oftenpresented as frequentist confidence intervals, and possibly on their own priors. For thesponsor, the optimal program design depends on its own prior and on the anticipatedreaction of other stakeholders. The questions to investigate are of the type: What is theprobability that the proposed design will lead to trial results that will convince regulatorsto approve the marketing of our drug and convince physicians to prescribe the drug?

Program 14.9 calculates expected utilities by integrating over the prior distribution forthe treatment effect δ (integration is performed using the QUAD function of SAS/IML).This prior distribution is assumed to be normal and its mean and standard deviation aregiven by the MUE and MUSE variables, respectively (notice that the case when MUSE=0corresponds to a model with a known treatment effect). SD is the residual standarddeviation. The value of MUE, SD, and some other parameters of the program are chosen tofacilitate the comparison with the approach described in Section 14.5.1. For example, theeffect size is centered around 0.2, the start-up cost is 50, and cost per patient is 0.1.

The program also allows for a possible non-inferior (but non-superior) result. If thisoption is not applicable, let NIMARGIN=0. The maximal possible value following anon-inferior result is given by the parameter VNIMAX and, similarly, VSUPMAX is themaximal value of a superior result.

In addition to the options of including a prior effect distribution and/or a non-inferioritycriterion, a time delay due to increased sample size may also be included. In the model, themaximal profit given marketing (VNIMAX or VSUPMAX) is decreased by a factorTIMEC times the sample size.

In the DO loop in Program 14.9, the expected utility (EUTILITY) is calculated for anumber of different sample sizes (given by the vector SIZE). On the negative side,TOT COST is the total cost for the trial and TIME FACTOR accounts for the relativeloss in value due to the time delay. PSUP and PNI are the probabilities for superiority andnon-inferiority, respectively. These are functions of the treatment effect. Given the effect,UTILITY is the expected gain, that is, probability-weighted profit corrected for the timedelay via TIME FACTOR, minus the total cost of running the trial. QUAD is applied tocalculate the expected utility over the prior distribution for the effect.

In the model underlying the program, the trial result is important only for the regulatorylabel (superior, non-inferior, or not approved). Given the label, the profit in this model isindependent of the trial result and sample size. If one is trying to apply decision analysison a practical Phase III sizing problem, we advise that the modeling of the profit be donewith greater care. If a better commercial model can be defined in terms of sample size andtrial results, it is often relatively straightforward to modify Program 14.9 accordingly.

Program 14.9 Expected utility when the treatment effect is uncertain

proc iml;muE=20;muSE=10;sd=100;NImargin=10;vSUPmax=1000;vNImax=400;alpha=0.05;crit=probit(1-alpha/2);timec=0.0001;startc=50;c=0.1;

Page 418: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 405

size=t(do(0,2000,10));m=nrow(size);Eutility=j(m,1,0);do k=1 to m;

n=size[k,1];time_factor=max(1-timec*n,0);tot_cost=startc+n*c;start h(effect) global(n,muE,muSE,sd,NImargin,crit,

vSUPmax,vNImax,time_factor,tot_cost);pSup=probnorm(effect/sd*sqrt(n/4)-crit);pNI=probnorm((effect+NImargin)/sd*sqrt(n/4)-crit)-pSup;utility=time_factor*(vSUPmax*pSup+vNImax*pNI)-tot_cost;density=exp(-(effect-muE)**2/(2*muSE**2))/sqrt(2*3.14159*muSE**2);h_out=density*utility;return(h_out);

finish;call quad(temp,"h",{.M .P}); /* Integrate over the real axis */Eutility[k,1]=temp;

end;create plotdata var{size Eutility};append;quit;

axis1 minor=none label=(angle=90 "Utility") order=(0 to 600 by 200);axis2 minor=none label=("Sample size") order=(0 to 2000 by 500);symbol1 i=join width=3 line=1 color=black;proc gplot data=plotdata;

plot Eutility*size/haxis=axis2 vaxis=axis1 frame;run;quit;

Figure 14.7 Expected utility as a function of sample size

Util

ity

0

200

400

600

Sample size

0 500 1000 1500 2000

The output of Program 14.9 is displayed in Figure 14.7. The figure depicts therelationship between the expected utility and total sample size. Under the assumptionsmade in the program, the optimal sample size is 887 patients, which is much smaller thanthe optimal sample size of 1431 patients for the model considered in Section 14.5.1.

Page 419: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

406 Pharmaceutical Statistics Using SAS: A Practical Guide

14.5.4 Do Not Sub-Optimize!A poor decision analysis, which ignores essential components of the problem, is often worsethan no formal decision analysis at all. As Einstein said, “Things should be made as simpleas possible — but no simpler”. We would therefore like to stress that the models presentedin Sections 14.5.1 and 14.5.3 must not be applied thoughtlessly to a practical problem.They should be seen just as examples of how a decision model can be built and refined andhow to analyze continuous decision problems using SAS. Additional factors which may beimportant in a practical situation are the demand for safety data, the relation between theamount of Phase III data and sales, alternative investment options in terms of other drugsin the pipeline, etc.

14.5.5 Integrating the ModelsMany problems involve a number of decisions, which have to be made at different times.Earlier decisions may lead to information that can be used as input for later decisions. Thisis obvious in clinical development where the final decisions concerning the design of thePhase III trials will be made after the analysis of a dose-finding trial (Phase IIb), whichmight follow upon a proof of principle trial (Phase IIa) and so forth. Such problems,involving sequential decisions, are often best attacked by backward induction (Bather,2000); the analysis starts by solving the decision problems at the latest stage and thenworks gradually backwards in time to reach an optimal solution of the entire sequentialproblem.

The drug development decision model of Sections 14.2 and 14.4, focusing on go/no godecisions, is naturally connected with the sample size decisions studied in the presentsection. Consider for a moment the integrated decision problem with the three questions:

• Should a pretest be run?• Should a Phase III trial be run?• What is the optimal sample size in a Phase III trial?

The best way to solve the composite problem is by starting to analyze the latest decision.The optimal sample size is determined for all the different scenarios, e.g.,

• Scenario 1: A positive pretest,• Scenario 2: A negative pretest,• Scenario 3: No pretest to be run,

and the corresponding conditional expected utility is calculated. Then, these values can beused as rewards in a decision tree that contains only the pretest and Phase III go/no godecisions. Similarly, PROC DTREE is working by backward induction and analyzes thelatest nodes first, taking expectation over chance nodes and maximizing over decisionnodes.

Yin (2002) analyses sample sizing in Phase II trials in light of a future go/no go decision.

14.6 Sequential Designs in Clinical TrialsSequential design and analysis plays a natural role in clinical trials, primarily because ofthe ability it gives of stopping early either if the treatment under test does not deliver theanticipated effect, or because it is more efficacious than was thought initially. Mostsequential approaches are based on frequentist statistics (see, for example, Armitage 1975and Whitehead 1997) and normally carried out group sequentially, as data are reviewedand monitored on a periodic basis. There is a long tradition of criticizing such frequentist

Page 420: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 407

approaches, firstly because by focusing on the consequences of the stopping rules on theType I error rate they contradict the likelihood principle (Anscombe, 1963; Cornfield,1969), and secondly because they ignore the fact that stopping is a decision whose lossesshould be taken into account.

There are a number of approaches that have been looked at to determine an optimaldecision rule in a sequential context. The first is traditionally the way that sequentialdecisions have been tackled and is based on backward induction (Berger, 1985).

The idea in backward induction is extremely simple and begins by considering the lastpossible decision that can be made at the point that the final data that could be gatheredhave been gathered. For every possible data set there is an optimal decision to be made.For example, in the case of the comparison of two treatments: Which is the best treatment,or Which can be recommended? Associated with the optimal decision is an expected loss inwhich the expectation is based on the posterior distribution of the parameters. Thepenultimate decision is considered next. Now there are three potential decisions that couldbe taken for each possible data set:

• Stop and recommend the first treatment.• Stop and recommend the second treatment.• Continue to the final stage.

If the trial is continued, the loss, which includes the cost of the sampling required to reachthe final decision point, can be determined, as can the expected losses of each decision tostop. Consequently, the optimal decision and its expected loss can be determined. Inprinciple, the process can be continued backwards in time to the first decision to be made.Such sequential decision problems are famously difficult because

. . . if one decision leads to another, then to analyse the former, one first needs toanalyse the latter, since the outcome of the former depends on the choice of the latter(French, 1989)

A consequence of which is that to completely solve the problem by backward inductioninvolves accounting for exponentially increasing numbers of potential scenarios (Berger,1985).

The second approach taken to solving sequential decision problems is a forwardalgorithm based on simulation. Before considering this approach, we will illustrate the ideaby using it to determine the optimal sample size as in Section 14.5.

Although the general decision problem formulated in Section 14.3 is seductively simple inits formality, its practical implementation is not always so simple. For complex models theintegrations required to determine the optimal Bayes’ decision may not be straightforwardparticularly in multi-parameter, non-linear problems or in sequential decision problems.

Even if the integrations necessary are analytically intractable, the Bayes solution caneasily be obtained by simulation. Recall that the Bayesian decision is obtained byminimizing ∫

X,θ

L(θ, d(X)) p(X | θ)π(θ) dθ

If the loss function L(θ, d(X)) is available for every combination of θ and d(X), thedecision problem can be solved as follows. Assuming that the generation of randomvariables from both π(X|θ) and p(θ) is possible, generate samples {θj , Xj}, j = 1, . . . , M ,and then evaluate for each simulation the loss, L(θj , d(Xj)). An estimate of the expected

Page 421: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

408 Pharmaceutical Statistics Using SAS: A Practical Guide

risk can be obtained from

1M

M∑j=1

L(θj , d(Xj)).

Numerical optimization of the resulting estimated expected risks gives the optimalBayesian decision.

In the context of finding optimal sample sizes for a clinical trial discussed inSections 14.5.1, this approach effectively corresponds to replacing the use of thePROBNORM function by simulation, which is hardly worthwhile. However, the sameapproach allows the simple relaxation of assumptions. For example, suppose that inProgram 14.6 we wish

• To use a t-test rather than a test based on a known variance.• To use proper prior information.

Simulation makes this very simple. Program 14.10 uses simulation to determine theoptimal sample size based on a t-statistic when prior information is available both for thetreatment effect (using a normal density) and the residual variance (using an inverse-χ2

density). Figure 14.8 plots the computed cost, gain, and utility functions.

Program 14.10 Simulating the optimal sample size using t-statistic and informative prior

data nsample;n_sim=1000; /* Number of simulations*/alpha=0.05; /* 2-sided significance level */pr_eff_m=35; /* Prior mean for treatment effect */pr_eff_s=100; /* Prior SD for treatment effect */pr_sd=100; /* Prior expected value of residual SD */pr_v=10; /* Number of df for prior inverse chi-square */g=1000; /* Value of a successful (i.e., significant) trial */startc=50; /* Start-up cost */c=1.0; /* Cost per patient */do n=10 to 1000 by 10;

t_df=n-2; critical=tinv(1-alpha/2,t_df);cost=startc+c*n;egain=0;do i_sim=1 to n_sim;

theta=pr_eff_m+pr_eff_s*normal(0);sigma_2=pr_v*pr_sd**2/(2*rangam(0,pr_v/2));y=theta+sqrt(sigma_2*4/n)*normal(0);t_statistic=y/(sqrt(sigma_2*4/n));if t_statistic>critical then egain=egain+g;

end;egain=egain/n_sim;utility=egain-cost;output;

end;axis1 minor=none label=(angle=90 "Utility") order=(-1000 to 1000 by 500);axis2 minor=none label=("Sample size") order=(0 to 1000 by 200);symbol1 i=spline width=3 line=1 color=black;symbol2 i=spline width=3 line=20 color=black;symbol3 i=spline width=3 line=34 color=black;

Page 422: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 409

proc gplot data=nsample;plot (cost egain utility)*n/overlay vaxis=axis1 haxis=axis2 frame;run;quit;

Figure 14.8 Cost (solid curve), expected gain (dashed curve), and utility (dotted curve) as a function of samplesize

Util

ity

-1000

-500

0

500

1000

Sample size

0 200 400 600 800 1000

Of course since we are using simulation techniques we need not be restricted to known,simple, convenient parametric forms for the priors. All that is required is an ability tosimulate from the priors.

14.6.1 Sequential Decision ProblemsTo illustrate how a simulation approach can be used to tackle some sequential decisionproblems we consider the following example based on the approach taken by Carlin et al.(1998). The context is a clinical trial in which the objective is to estimate the treatmenteffect θ of a new active therapy (A) relative to placebo (P). Large negative values aresuggestive of superiority of A over P and conversely large positive values favor P. Theprotocol allows for a single interim analysis. Along the lines of Freedman and Spiegelhalter(1989), suppose that an indifference region (c2, c1) is predetermined such that

• A is preferred if θ < c2.• P is preferred if θ > c1.• Either treatment is acceptable if c2 < θ < c1.

At the final analysis there are two decisions available: d1 to decide in favor of A and d2to decide in favor of P. For each decision, the loss functions given the true value of θ are:

l1(d1, θ) = s1(θ − c1), l2(d2, θ) = s2(c2 − θ),

where s1 and s2 are positive constants. These losses are negatively increasing (gains) forcorrect decisions. Given the posterior at the final analysis, p(θ | X1, X2), the best decision isdetermined by the smaller of the posterior expected losses:

E[l1(d1, θ) | X1, X2] = s1(E[θ | X1, X2] − c1),

E[l2(d2, θ) | X1, X2] = s2(c2 − E[θ | X1, X2]).

Page 423: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

410 Pharmaceutical Statistics Using SAS: A Practical Guide

By equating these losses and solving for E[θ | X1, X2], it is clear that Decision d1 will beoptimal if

E[θ | X1, X2] ≤ s1c1 + s2c2

s1 + s2.

Otherwise Decision d2 is optimal.At the interim analysis, in addition to Decisions d1 and d2 to stop in favor of A or P,

respectively, based on p(θ | X1), there is a further decision possible, which is to proceed tothe final analysis, to incur the extra cost (c3) of the patients recruited between the interimand final analyses and then to make the decision.

If σ2 is known, the forward approach to the decision is algorithmically as follows:

1. Based on the prior distribution and the first stage data X1, determine the posteriordistribution p(θ | X1, σ

2).2. Determine the expected losses s1(E[θ | X1] − c1) and s2(c2 − E[θ | X1]).3. Let k be the number of simulatons. For i = 1 to k,

(a) Simulate θi from p(θ | X1, σ2).

(b) Simulate data X2,i from p(X2 | θi, σ2).

(c) Calculate s1(E[θ | X1, X2,i] − c1) and s2(c2 − E[θ | X1, X2,i]).4. Let lfinal be the minimum of the following two quantities:

1k

k∑i=1

s1(E[θ | X1, X2,i] − c1),1k

k∑i=1

s2(c2 − E[θ | X1, X2,i]).

5. Choose the minimum of

s1(E[θ | X1] − c1), s2(c2 − E[θ | X1]), and c3 + lfinal.

Obvious modifications need to be made if σ2 is unknown.Implementation of this approach is shown in Program 14.11. The output of the program

(Figures 14.9 and 14.10) defines regions for the observed treatment difference at theinterim for which it is preferable to stop rather than to go on to collect more data andconduct the final analysis.

Program 14.11 Computation of loss functions in the Bayesian sequential design problem

data interim_decision;n_sim=10000; /* Number of simulations*/s1=1; /* Cost associated with falsely choosing d1 (active)*/

s2=1; /* Cost associated with falsely choosing d2 (placebo)*/s3=0.1; /* Cost associated with sampling after interim*/n1=1; /* Sample size per group at interim */n2=1; /* Sample size per group after interim */

sigma_2=0.5**2;/* Known sigma**2 */c1=0; /* Upper indifference boundary (gt favours placebo)*/c2=-0.288; /* Lower indifference boundary (lt favours treatment)*/

pr_m=0.021; /* Prior mean */pr_sd=0.664; /* Prior standard deviation */

/* Looping over an observed treatment effect (n1 patients per group) at the interim */do x1=-1 to 1 by 0.02;

Page 424: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 411

/* Determine posterior mean (interim) and standard deviation*/

posterior_m_1=(x1*pr_sd**2+2*pr_m*sigma_2/n1)/(pr_sd**2+2*sigma_2/n1) ;posterior_sd_1=sqrt(1/(1/pr_sd**2+n1/(2*sigma_2)));/* Determine losses at interim (loss_11: decision 1, loss_21: decision 2) */

loss_11=max(0,s1*(posterior_m_1-c1));loss_21=max(0,s2*(c2-posterior_m_1));/* Determine minimum of losses at interim */loss_interim=min(loss_11,loss_21);

/* Initialise summation variables for future decisions */loss_12_sum=0; loss_22_sum=0;

/* Simulate future data */do i_sim=1 to n_sim;

/* Simulate treatment parameter from posterior at interim */theta=posterior_m_1+posterior_sd_1*normal(0);/* Simulate treatment effect from n2 patients on each treatmentfor given theta */

x2=theta+sqrt(sigma_2*2/n2)*normal(0);/* Determine posterior mean (final) and standard deviation*/posterior_m_2=(x2*posterior_sd_1**2+2*posterior_m_1*sigma_2/n2)/

(posterior_sd_1**2+2*sigma_2/n2);/* Determine losses at final (loss_12: decision 1, loss_22: decision 2) */loss_12=s3+max(0,s1*(posterior_m_2-c1));

loss_22=s3+max(0,s2*(c2-posterior_m_2));/* Accumulate losses */loss_12_sum=loss_12_sum+loss_12;loss_22_sum=loss_22_sum+loss_22;

end;

/* Calculate expected losses */

loss_12=loss_12_sum/n_sim; loss_22=loss_22_sum/n_sim;/* Determine minimum of losses at final */loss_final=min(loss_12,loss_22);

output;end;keep loss_11 loss_21 loss_12 loss_22 loss_final loss_interim x1;

axis1 minor=none label=(angle=90 "Loss") order=(0 to 0.6 by 0.2);axis2 minor=none label=("First stage data (X1)") order=(-1 to 1 by 0.5);symbol1 i=spline width=3 line=20 color=black;symbol2 i=spline width=3 line=34 color=black;

symbol3 i=spline width=3 line=1 color=black;/* Plot loss functions for interim analysis */proc gplot data=interim_decision;

plot (loss_11 loss_21 loss_interim)*x1/overlay vaxis=axis1 haxis=axis2 frame;run;

/* Plot loss functions for final analysis */

proc gplot data=interim_decision;plot (loss_12 loss_22 loss_final)*x1/overlay vaxis=axis1 haxis=axis2 frame;run;quit;

In practice, Carlin et al. (1998) use this approach to define a series of critical values forthe posterior mean of the treatment effect for each of a number of interims. The advantageof their approach is that the critical values can be determined a priori and there is no needfor complex Bayesian simulation during the course of the study. Kadane and Vlachos(2002) develop an alternative approach intended, in their words, to “capitalize on thestrengths, and compensate for the weakness of both the backwards and forward strategies”.

Page 425: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

412 Pharmaceutical Statistics Using SAS: A Practical Guide

Figure 14.9 Loss functions for Decision 1 (dashed curve), Decision 2 (dotted curve), and minimum loss function(solid curve) at the interim analysis

Loss

0.0

0.2

0.4

0.6

First stage data (X1)

-1.0 -0.5 0.0 0.5 1.0

Figure 14.10 Loss functions for Decision 1 (dashed curve), Decision 2 (dotted curve), and minimum lossfunction (solid curve) at the final analysis

Loss

0.0

0.2

0.4

0.6

First stage data (X1)

-1.0 -0.5 0.0 0.5 1.0

They develop a hybrid algorithm that works backwards as far as is feasible, determiningthe expected loss of the appropriate optimal continuation as a “callable function”. It thenproceeds using the forward algorithm from the trial’s start to where the backwardalgorithm becomes applicable. This reduces the size of the space to be covered and hencewill increase efficiency. Berry et al. (2000) describe a similar strategy in the context of adose-response study.

14.7 Selection of an Optimal DoseOne of the most crucial decisions in the development of a new drug is which dose to use.The situation varies between different indications. Sometimes the dose can be titrated foreach patient, sometimes the dose is individualized based on gender, body weight, geneticconstitution, and other factors. Often one looks for a one-dose-fits-all. The optimal dose isa compromise between beneficial effects and undesired side effects. In general, both effectsand, especially, side effects, may be multi-dimensional. As a further complication, whichside effects are important may not be clear until after the Phase III program or even after

Page 426: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 413

marketing of the drug. We choose to treat only the simple situation with a one-dimensionaleffect variable and a one-dimensional, well-specified type of adverse event. A practicalexample of finding the optimal dose, based on decision analysis and modeling of efficacyand safety, is provided by Graham et al. (2002).

Effects and side-effects are typically measured on different scales and defining theoptimal dose requires that they be weighted together in some way. It is, however, notobvious how to weigh a decrease in blood pressure against a risk of severe coughing, or howto weigh a decrease in body weight against problems with loose stools. Although suchweighting has to be subjective, there is no rational way to avoid it. Given dose-responseinformation for effects and adverse events, to choose a dose more or less implies at least animplicit statement about the relative importance of the two dimensions.

On some occasions, it may occur that effects and side effects can be measured directlyon the same scale. One example is when both the effect and the side effect are measured interms of living and dying. Even though effects and side effects appear to be measured onthe same scale, it is not obvious that they should be weighted equally. Different causes ofdeath may give different opportunities for the patient to prepare for death. Even if theoutcomes are identical in almost everything, it may subjectively be harder to acceptnegative events due to the treatment than similar negative events due to the naturalpropagation of the disease. Furthermore, there may be a legal difference: it is easier toblame the doctor for a treatment’s adverse effects than for the negative effects of theuntreated disease. Thus, a weighting of adverse effects relative beneficial effects is generallyneeded.

Suppose that the effect is a continuous variable and the adverse effect is dichotomous.Let E(D) be the expected effect and p(D) the probability that a patient experiences anAE, as functions of the dose D. One reasonable utility function is the simple weightedcombination

U(D) = E(D) − kp(D).

Since it is often hard to choose a value of the weight k, it is useful to investigate therobustness of the conclusions over a range of values of k.

A common model for the effect is the so-called Emax model

E(D) = E0 + Emax · Dγ

Dγ + EDγ50

. (14.1)

The parameters E0, Emax and ED50 have natural interpretations as placebo response, themaximal possible placebo-adjusted effect and the dose for which the adjusted effect is 50%of Emax, respectively. The Hill coefficient γ is related to the steepness of the dose-responsecurve around ED50.

Knowledge of the effect curve alone does not imply what the optimal dose is; safetyinformation is also needed. Assume a logistic model for the probability of AE as a functionof the logarithm of the dose,

p(D) =exp(α + β log(D))

1 + exp(α + β log(D)).

14.7.1 Models without UncertaintyFirst, assume all parameters to be fixed. Here is an example:

E0 = 0, Emax = 300, ED50 = 0.3, γ = 1.0, α = −1.5, β = 2.0.

Assume also that the weight k = 1000, meaning that 0.1% AEs is equalized to one unit ofeffect. Program 14.12 examines the relationship between the dose of an experimental drugD and the expected effect E(D), adverse event probability p(D), and utility U(D).

Page 427: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

414 Pharmaceutical Statistics Using SAS: A Practical Guide

Program 14.12 Plot of the dose-effect, dose-Prob(AE), and dose-utility functions

/* Define model parameters and macros */%let k=1000;%let emax=300;%let ed50=0.3;%let gamma=1.0;%let e0=0.0;%let alpha=-1.5;%let beta=2.0;%macro effmodel(dose,emax,ed50,gamma,e0);

%global effout;%let effout=&e0+&emax*&dose**&gamma/(&dose**&gamma+&ed50**&gamma);

%mend effmodel;%macro AEmodel(logdose,alpha,beta);

%global AEout;%let AEout=exp(&alpha+&beta*&logdose)/(1+exp(&alpha+&beta*&logdose));

%mend AEmodel;/* Create data set for plotting */data window;

do logd=log(0.01) to log(10.0) by log(10.0/0.01)/100;dose=exp(logd);%effmodel(dose=dose,emax=&emax,ed50=&ed50,gamma=&gamma,e0=&e0);effect=&effout;%AEmodel(logdose=logd,alpha=&alpha,beta=&beta);AEprob=&AEout;AEloss=&k*AEprob;utility=effect-AEloss;output;

end;axis1 minor=none label=(angle=90 "Utility") order=(-300 to 300 by 100);axis2 minor=none label=("Dose") logbase=10 logstyle=expand;symbol1 i=join width=3 line=1 color=black;symbol2 i=join width=3 line=20 color=black;symbol3 i=join width=3 line=34 color=black;proc gplot data=window;

plot (effect AEloss utility)*dose/haxis=axis2 vaxis=axis1overlay frame vref=0 lvref=34;run;quit;

Figure 14.11 displays the dose-effect, dose-Prob(AE), and dose-utility functions. It isclear that the first two functions, E(D) and p(D), are monotone functions of the dose. Bycontrast, the dose-utility function is increasing on [0, D∗] and decreasing on [D∗, 10], whereD∗ is the dose that maximizes the utility function. The optimal dose, D∗, is easy to findusing PROC NLP (see Program 14.13).

Program 14.13 Computation of the optimal dose

proc nlp outest=result noprint;max utility;decvar dose;%effmodel(dose=dose,emax=&emax,ed50=&ed50,gamma=&gamma,e0=&e0);effect=&effout;%AEmodel(logdose=log(dose),alpha=&alpha,beta=&beta);AEprob=&AEout;utility=effect-&k*AEprob;

Page 428: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 415

Figure 14.11 Plot of the dose-effect (solid curve), dose-Prob(AE) (dashed curve), and dose-utility (dottedcurve) functions

Util

ity

-300

-200

-100

0

100

200

300

Dose

0.01 0.10 1.00 10.00

data result;set result;format optdose effect utility 6.2 AEprob 5.3;where _type_="PARMS";optdose=dose;%effmodel(dose=optdose,emax=&emax,ed50=&ed50,gamma=&gamma,e0=&e0);effect=&effout;%AEmodel(logdose=log(optdose),alpha=&alpha,beta=&beta);AEprob=&AEout;utility=effect-&k*AEprob;label optdose="Optimal dose"

effect="Effect"AEprob="Probability of an AE"utility="Utility";

keep optdose effect AEprob utility;proc print data=result noobs label;

run;

Output from Program 14.13

Optimal Probabilitydose Effect Utility of an AE

0.42 175.02 137.13 0.038

Output 14.13 lists the optimal dose, expected response, utility and, finally, probabilityof observing an AE at this dose. The optimal dose is D∗ = 0.42, and the associated netutility is given by

U(D∗) = E(D∗) − kp(D∗) = 175 − 1000 × 0.038 = 137.

14.7.2 Choosing the Optimal Dose Based on Limited DataIn reality, the efficacy and safety models considered above are uncertain and have to beestimated from the information at hand. We will assume that a relatively smalldose-finding trial (124 patients) has been run to study the efficacy and safety of five doses

Page 429: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

416 Pharmaceutical Statistics Using SAS: A Practical Guide

of an experimental drug. The data collected in the study (DOSE, EFF, and AE variables)are contained in the STUDY data set available on the book’s companion Web site. The AEvariable is 1 for patients who have experienced the AE, and 0 otherwise. The EFF variableis the observed effect. Table 14.1 displays the summary of the efficacy and safety data inthe STUDY data set.

Table 14.1 Summary of the Efficacy and Safety Data in theDose-Finding Trial (STUDY Data Set)

Efficacy variableDose

Number ofpatients

Number ofAEs Mean SD

0.03 26 0 −13.7 291.80.1 23 0 135.6 269.30.3 25 1 132.6 267.01 26 5 293.0 328.33 24 10 328.4 339.1

In addition to this limited information on the new drug’s effect, there is a lot ofliterature data for other drugs from the same class. Based on this, we think that we canestimate the E0, Emax, and γ parameters of the Emax model, introduced earlier in thissection, sufficiently well. (It is assumed that the parameters are shared by all drugs in theclass.) Thus, we will take E0 = 0, Emax = 300, and γ = 1.0 as fixed. Only the potency,described by ED50, of our new drug remains uncertain. The computations are simplified, asonly one of the four parameters in the Emax model must be estimated based on the smallin-house trial.

Before estimating the unknown parameters, ED50, α, and β, it is good to consider howreliable the efficacy and safety models are. Even if the models fit the data well, we shouldnot be over-confident. For example, it is often possible to fit logit, probit, andcomplementary log-log models to the same data set. Still, these models have a largerelative difference in the predicted probability of AE at very low doses. In our example, asin many practical situations, there is not enough data to discriminate between differentreasonable types of models. A model-independent analysis is therefore a good start.

Program 14.14 calculates estimates (EFF MEAN, UTIL EST, P EST) and theirstandard errors (EFF SE, UTIL SE, P SE) for mean effect, mean utility, and probability ofan AE, respectively, in each of the five dose groups. Note that, depending on the nature ofthe effect and AE variables, it is plausible that they are positively or negatively correlated.For the sake of simplicity, however, we assume independence throughout this section. Thisassumption may alter the estimated standard error for the utility but is unlikely to have amajor impact on the estimated mean utility.

Program 14.14 Model-free estimates and SEs of E(D), p(D), and U(D)

%let k=1000; /* Weighting coefficient */proc means data=study.study;

class dose;var eff;output out=summary_eff(where=(_type_=1)) n=n mean=mean std=std;

proc freq data=study.study;table dose*ae/out=summary_ae(where=(ae=1));

data summary;merge summary_eff summary_ae;by dose;if count=. then count=0;n_ae=count;

Page 430: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 417

eff_mean=mean;eff_sd=std;keep dose n n_ae eff_mean eff_sd;

data obs_util;set summary;format eff_mean eff_se util_est util_se 5.1 p_est p_se 5.3;eff_se=eff_sd/sqrt(n);p_est=n_ae/n;p_se=sqrt(p_est*(1-p_est)/n);util_est=eff_mean-&k*p_est;util_se=sqrt(eff_se**2+(&k*p_se)**2);label dose="Dose"

eff_mean="Effect"eff_se="Effect (SE)"p_est="Probability of an AE"p_se="Probability of an AE (SE)"util_est="Utility"util_se="Utility (SE)";

keep dose eff_mean eff_se p_est p_se util_est util_se;proc print data=obs_util noobs label;

run;

Output from Program 14.14

ProbabilityEffect Utility Probability of an AE

Dose Effect (SE) Utility (SE) of an AE (SE)

0.03 -13.7 57.2 -13.7 57.2 0.000 0.0000.10 135.6 56.2 135.6 56.2 0.000 0.0000.30 132.6 53.4 92.6 66.2 0.040 0.0391.00 293.0 64.4 100.7 100.6 0.192 0.0773.00 328.4 69.2 -88.3 122.1 0.417 0.101

Output 14.14 lists the three estimated quantities (mean effect, mean utility, andprobability of an AE) along with the associated standard errors. The highest observedutility is for dose 0.1. For this dose, the utility is significantly better than placebo (as thez-score is 135.6/56.2 ≈ 2.42). Considering the large standard errors, however, it is hard todistinguish between doses in a large range. The only immediate conclusion is that theoptimal dose should be higher than 0.03 and probably lower than 3.

After using a model-free approach, we proceed to fit the Emax model to the efficacy dataand logistic model to the safety data. Program 14.15 uses the NLIN and LOGISTICprocedures, respectively, to calculate parameter estimates in the two models. Theparameter estimates are assigned to macro variables for use later.

Program 14.15 Estimating parameters and posterior distribution

data studylog;set study;logdose=log(dose);

%let e0=0;%let emax=300;%let gamma=1.0;

Page 431: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

418 Pharmaceutical Statistics Using SAS: A Practical Guide

proc nlin data=study outest=estED50;ods select ParameterEstimates;parms log_ed50=-1;model eff=&e0+&emax*dose**&gamma/(dose**&gamma+exp(log_ed50)**&gamma);output out=Statistics Residual=r parms=log_ed50;

data estED50;set estED50;if _type_="FINAL" then

call symput(’log_est’,put(log_ed50, best12.));if _type_="COVB" then

call symput(’log_var’,put(log_ed50, best12.));run;

proc logistic data=studylog outest=est1 covout descending;ods select ParameterEstimates CovB;model ae=logdose/covb;

data est1;set est1;if _type_=’PARMS’ then do;

call symput(’alpha_mean’,put(intercept, best12.));call symput(’beta_mean’,put(logdose, best12.));

end;else if _name_=’Intercept’ then do;

call symput(’alpha_var’,put(intercept, best12.));call symput(’ab_cov’,put(logdose, best12.));

end;else if _name_=’logdose’ then call symput(’beta_var’,put(logdose, best12.));run;

Output from Program 14.15

The NLIN Procedure

Approx Approximate 95% ConfidenceParameter Estimate Std Error Limits

log_ed50 -1.5794 0.5338 -2.6360 -0.5228

The LOGISTIC Procedure

Analysis of Maximum Likelihood Estimates

Standard WaldParameter DF Estimate Error Chi-Square Pr > ChiSq

Intercept 1 -1.6566 0.3435 23.2639 <.0001logdose 1 1.2937 0.3487 13.7647 0.0002

Estimated Covariance Matrix

Variable Intercept logdose

Intercept 0.117968 -0.05377logdose -0.05377 0.1216

Output 14.15 lists the estimated model parameters. Note that a direct estimation ofED50 (not shown) will result in a 95% confidence interval which contains negative values.Such values are not plausible. As the uncertainty in the parameter estimates will be

Page 432: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 419

important later, we prefer to estimate log(ED50) rather than ED50. The estimate andsample standard error of log(ED50) are −1.5794 and 0.5338, respectively.

The estimates of α and β in the logistic model for the probability of observing an AEare α = −1.6566 and β = 1.2937, respectively. The covariance matrix is governed by

Σ =[

0.118 −0.054−0.054 0.122

].

One obvious way to consider the parameter uncertainty is to apply a Bayesian viewpointand consider the posterior distributions for the parameters. The posterior distribution isproportional to the prior multiplied by the likelihood. PROC LOGISTIC and PROC NLINare likelihood-based procedures and the output from them can be used to get at leastrough approximations of the likelihood functions. Assuming that the priors are relativelyflat, we may take the two-dimensional normal distribution with mean[

α

β

]and covariance matrix Σ as the approximate posterior for[

αβ

].

Similarly, the estimate of log(ED50) and its standard error may, to a reasonableapproximation, serve as the mean and standard deviation in a normal prior for log(ED50).

Program 14.16 uses the previously calculated macro variables to simulate utility curvesbased on the posteriors for the parameters. Figure 14.12 displays a number of simulatedcurves together with the simulated expected utility curve.

Program 14.16 Estimating parameters and posterior distribution

%let nsim=1000; /* Number of simulated curves */%let ndisplay=10; /* Number of displayed individual curves */%let doseint=100; /* Dissolution of dose scale */%macro out_sim;

%do __cnt=1 %to &ndisplay.;simu&__cnt.=t(util_sim[&__cnt.,]);

%end;create simdata var{dose logdose Eutility

%do __cnt=1 %to &ndisplay.; simu&__cnt. %str( ) %end;};append;

%mend out_sim;%macro ind_plot;

%do __cnt=1 %to &ndisplay.;simu&__cnt.*dose

%end;%mend ind_plot;/* Simulations */proc iml;

seed=257656897;z=normal(repeat(seed,&nsim,3));alpha_sd=sqrt(&alpha_var);beta_sd=sqrt(&beta_var);ab_corr=&ab_cov/sqrt(&alpha_var*&beta_var);alpha_sim=&alpha_mean+alpha_sd*z[,1];beta_sim=&beta_mean+beta_sd*(ab_corr*z[,1]+sqrt(1-ab_corr**2)*z[,2]);logED50_sd=sqrt(&log_var);logED50_sim=&log_est+logED50_sd*z[,3];ED50_sim=exp(logED50_sim);

Page 433: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

420 Pharmaceutical Statistics Using SAS: A Practical Guide

logdose=t(do(log(0.01),log(10),log(10/0.01)/&doseint));logdosematrix=j(&nsim,1)*t(logdose);dose=exp(logdose);dosematrix=exp(logdosematrix);onevector=j(1,nrow(logdose));alpha_sim_matrix=alpha_sim*onevector;beta_sim_matrix=beta_sim*onevector;ED50_sim_matrix=ED50_sim*onevector;logit=exp(alpha_sim_matrix+beta_sim_matrix#logdosematrix);prob_sim=logit/(1+logit);Eprob=t(j(1,&nsim)*prob_sim/&nsim);effect_sim=&e0+&emax*dosematrix##&gamma/

(dosematrix##&gamma+ED50_sim_matrix##&gamma);Eeffect=t(j(1,&nsim)*effect_sim/&nsim);

util_sim=effect_sim-&k*prob_sim;Eutility=t(j(1,&nsim)*util_sim/&nsim);/* Create data set containing simulation results */%out_sim;quit;

/* Display mean curve and &ndisplay individual simulation curves */axis1 minor=none label=(angle=90 "Utility") order=(-600 to 200 by 200);axis2 minor=none label=("Dose") logbase=10 logstyle=expand;symbol1 i=join width=5 line=1 color=black;symbol2 i=join width=1 color=black line=34 repeat=&ndisplay.;proc gplot data=simdata;

plot Eutility*dose %ind_plot/overlay vaxis=axis1 haxis=axis2 vref=0 lvref=34;run;quit;

Figure 14.12 Plot of simulated utility functions (dashed curves) and simulated expected utility function (solidcurve)

Util

ity

-600

-400

-200

0

200

Dose

0.01 0.10 1.00 10.00

When one single dose has to be chosen, the expected utility is a good criterion. Thevariability between the different simulated utility curves is, however, of great interestespecially when other options are possible. If the variability in optimal dose is small, thereis little reason to proceed with more than one dose. However, if the variability in utilitybetween doses that may be optimal is considerable, then there are arguments to bring twoor more doses to the next phase of investigations. In our example, the optimal dose is likely

Page 434: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 421

to be around 0.4, with limited uncertainty. However, recall that this analysis is dependenton a number of assumptions, such as the logistic and Emax models, normal approximationsof the posterior, and a flat prior. In practical work, it is important to think about thereliability of the assumptions and to assess the robustness against possible deviations fromthe assumptions. For example, the optimal dose may be plotted over a range of possibleweight coefficients k.

The population model used in Program 14.16 may be refined. Individuals differ in theirresponse to treatment. One reason is pharmacokinetic variability. This can be modeled andsometimes used to improve the benefit-risk relation by individualizing the dose.

14.8 Project PrioritizationAn important set of decisions with which a pharmaceutical sponsor is faced is that of thechoice of drug development projects and more generally of the prioritization of the projectschosen. This topic has been considered by a number of authors, in particular by Bergmanand Gittins (1985) but also by Senn (1996, 1997, 1998), Senn and Rosati (2002), Zipfel(2003), and Burman and Senn (2003).

It is instructive to consider the simpler problem of deciding which drugs to develop,since assessing the value of potential projects prior to inclusion in a portfolio may alsosuggest how, if at all, those projects that are included might be prioritized. A simpleanalogy, which is frequently made, is that of packing a number of objects of different valuesand volumes into a suitcase so that the resulting package is as valuable as possible. Thesuitcase is a metaphor for the constraints facing drug developers, the volumes of theobjects the extent to which the projects contribute individually to reaching or exceedingthe constraints and the values of the objects as the expected returns the objects will bringto the sponsor. Put like this, the problem is one of optimization subject to constraints, andit might be thought that linear programming, more specifically integer programming,would be the appropriate tool for solving the problem. In fact, there are various aspects ofthe problem that suggest that these techniques may be less useful than might at first sightbe supposed (this point will be taken up later) and in any case it is useful to examineindividual projects in terms of return on investment. We will review this aspect first beforereturning to optimization considerations.

The simplest useful index that may be used to evaluate projects in development for thepurpose of portfolio management is the so-called Pearson Index (Pearson, 1972), which is,effectively, a return on investment index. This calculates for a given project the expectedreward as a ratio of the expected cost. It may be defined as follows. First, we assume thatthe project consists of n stages that have been optimally sequenced. Sunk costs are to beignored, so that at any stage we can redefine a project, for the purpose of making decisions,as consisting of future stages only. The discounted cost of a given stage i is ci and theconditional probability of success in that stage given that the project has reached thatstage is pi, with p0 = 1 by convention. The expected current value of the future revenue ofa successful project is r. Then the Pearson Index may be written as

PI =r∏n

i=1 pi −∑n

i=1 ci

∏i−1j=0 pi∑n

i=1 ci

∏i−1j=0 pi

Here the denominator is the expected future cost of a project and this reflects the factthat costs will be paid only for a given stage if it is reached and not if the project fails. Thefirst term in the numerator is the unconditional expected future revenue. That is, it is theexpected future revenue, r, of a successful project multiplied by the probability of success,∏n

i=1 pi. Subtracting the denominator from this term produces the expected return net ofexpected costs, which is then the numerator of the Pearson Index.

Page 435: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

422 Pharmaceutical Statistics Using SAS: A Practical Guide

The index has the necessary property that other things being equal it will rank projectswith a favorable cost and probability architecture (Senn, 1996, 1998; Zipfel, 2003) morehighly than those with an unfavorable architecture. Other things being equal, projects thatwill fail early (if they fail) are to be preferred to those that will fail late and projects withlater costs are to be preferred to those with earlier ones. An example is given by Senn(1996).

Consider four projects, A, B, C, and D in three stages, details for which (including costsand success probabilities) are given in Table 14.2. The value of r (reward) in all four casesis 28. There is nothing to choose among these projects in terms of cost to see to completion(1 + 2 + 3 = 6), nor in terms of return if successful (28 − 6 = 22), nor in terms ofprobability of success (0.8 × 0.6 × 0.4 = 0.192). These figures are the same in all four cases.However, Projects C and D have a favorable cost architecture, and Projects B and D havea favorable probability architecture. In fact, as shown in Table 14.2, the Pearson Indexvalues for the four projects are 0.058 for Project A, 0.331 for Projects B and C, and 1.133for Project D. The index indicates a ratio of 1.133/0.058 = 19.5 between Projects D and Aand thus the most attractive project (Project D) is nearly 20 times as attractive as theleast attractive one (Project A).

Table 14.2 Costs and Success Probabilities for Four Hypothetical Projects

ProjectA B C D

Cost ($100m) Stage 1 3 3 1 1Stage 2 2 2 2 2Stage 3 1 1 3 3Total 6 6 6 6

Success Stage 1 0.8 0.4 0.8 0.4probability Stage 2 0.6 0.6 0.6 0.6

Stage 3 0.4 0.8 0.4 0.8Overall 0.192 0.192 0.192 0.192

Reward ($100m) 28 28 28 28

Expected cost 5.1 4.0 4.0 2.5Expected reward 5.4 5.4 5.4 5.4Expected profit 0.3 1.3 1.3 2.9Pearson Index 0.058 0.331 0.331 1.133

The three-stage example of the Pearson Index is simple enough to be solved with a handcalculator. In a more complex situation, PROC DTREE may, of course, be useful.Program 14.17 calculates the expected reward, i.e., the numerator in the Pearson Index, fortwo of the scenarios. The original PEARSON data set gives the cost architecture forProject A (and B). It shows that the costs of the three successive trials are in order 1, 2,and 3, and that the tree reaches the end as soon as a trial failure is encountered. It alsogives the terminal gain 28 in case all the chance events turn out to be OK trial results.

The MODIFY statement in PROC DTREE can be used interactively to modify thecosts for the different stages. In the program, the cost of the first trial is changed from 1 to3 and for the third trial from 3 to 1. The resulting PEARSON data set contains theappropriate costs for trial C (and D). The PROB data set states that the probabilities offailure are 0.2, 0.4, and 0.6 for Trials 1, 2, and 3, respectively. This is the probabilityarchitecture for Projects A and C.

Page 436: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 423

Program 14.17 Calculaton of the expecting reward in the Pearson model

data pearson;input _stname_ $ 1-8 _sttype_ $ 9-12 _outcome_ $ 13-22

_reward_ 23-26 _success_ $ 27-36;datalines;

Start D No go . .. Go -1 Stage1

Stage1 C Failure1 . .. OK1 -2 Stage2

Stage2 C Failure2 . .. OK2 -3 Stage3

Stage3 C Failure3 . .. OK3 28 .

;data prob;

input _event1_ $10. _prob1_ _event2_ $5. _prob2_;datalines;

Failure1 0.2 OK1 0.8Failure2 0.4 OK2 0.6Failure3 0.6 OK3 0.4

;proc dtree stagein=pearson probin=prob;

evaluate/summary;save;modify Go reward -3;modify OK2 reward -1;evaluate/summary;run;quit;

Output from Program 14.17

Decision Parameters

Decision Criterion: Maximize Expected Value (MAXEV)Optimal Decision Yields: 1.34

Optimal Decision Policy

Up to Stage Start

Alternatives Cumulative Evaluatingor Outcomes Reward Value----------------------------------------No go 0 0.00Go -1 2.34*

Decision Parameters

Decision Criterion: Maximize Expected Value (MAXEV)Optimal Decision Yields: 0.296

Page 437: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

424 Pharmaceutical Statistics Using SAS: A Practical Guide

Optimal Decision Policy

Up to Stage Start

Alternatives Cumulative Evaluatingor Outcomes Reward Value----------------------------------------No go 0 0.000Go -3 3.296*

The way that the Pearson Index could be used in practice would be to rank projects bytheir ratios and to select them starting with the highest and proceeding as far down the listas constraints permit. This is strongly analogous to the Neyman-Pearson lemma whichindicates that selecting points in the sample space to construct a critical region such that adesideratum of power is maximized subject to a constraint of size (Type I error rate)should be done by including points with the highest likelihood ratio1. For the projectselection problem, the overall research budget is analogous to the tolerated Type I errorrate, the expected return is analogous to power, and the Pearson Index to the likelihoodratio. This brings us back to the issue of integer programming discussed previously. As iswell known in the context of hypothesis testing, for problems in which the sample space isdiscrete, improved power can be delivered for a given size under suitable circumstances bygoing beyond the likelihood ratio criterion. The apparently unending discussion of“superior” alternatives to Fisher’s exact test bears witness to this. The analogy for theportfolio selection problem would be that by going beyond the Pearson Index formulationwe might, by sacrificing a large project with a high index but replacing it by two smallerprojects with less favorable indices, end up with a more valuable portfolio. This is,however, probably not a good idea for similar reasons to those that suggest that“improvements” to Fisher’s exact test should not be employed. It is better to look uponthe resources available in a drug development project as having some flexibility so that bytrading projects or raising capital one could avoid such index-violating decisions.

Nevertheless, for the sake of completeness and in order to illustrate further features ofSAS, we will demonstrate the application of integer programming in a simple examplebelow. This should not be taken as an indication that this is necessarily the way we believethat the portfolio management problem should be approached.

Let us look at a simple portfolio planning example using integer programming(Program 14.18). The costs and rewards for ten project candidates are specified in thePROJECTS data set. The budget restriction is given by the RHS variable and set to 100.Finally, all optimization variables are binary. That is, a project is either run (ACTIVE=1)or not run (ACTIVE=0).

Program 14.18 Integer programming to optimize portfolio

data projects;input _id_ $6. project1-project10 _type_ $ _rhs_;datalines;

cost 47 35 22 22 18 16 15 10 9 6 le 100reward 95 75 40 25 22 20 46 15 30 32 max .binary 1 1 1 1 1 1 1 1 1 1 binary .;

1It is perhaps worth pointing out that Alan Pearson of the Pearson Index is not to be confused with Egon Pearson of theNeyman-Pearson lemma!

Page 438: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 425

proc lp data=projects activeout=results;proc transpose data=results out=results;proc sort data=results;

by _name_;proc transpose data=projects out=projects;

id _id_;proc sort data=projects;

by _name_;data together (rename=(_name_=Projects));

merge projects results;by _name_;activity=col1;index=reward/cost;if substr(_name_,1,7)="project";keep _name_ cost reward index activity;

proc sort data=together;by descending index;

proc print data=together;run;

Output from Program 14.18

The LP Procedure

Constraint Summary

Constraint S/S DualRow Name Type Col Rhs Activity Activity

1 cost LE 11 100 99 02 reward OBJECTVE . 0 243 .

Obs Projects cost reward activity index

1 project10 6 32 1 5.333332 project9 9 30 1 3.333333 project7 15 46 1 3.066674 project2 35 75 0 2.142865 project1 47 95 1 2.021286 project3 22 40 1 1.818187 project8 10 15 0 1.500008 project6 16 20 0 1.250009 project5 18 22 0 1.2222210 project4 22 25 0 1.13636

Output 14.18 displays some of the output from the LP procedure as well as a list of theprojects, ordered by the value of the reward/cost-index, and indicating the optimal set ofprojects. The output of PROC LP states that the optimal solution gives a reward of 243 ata cost of 99. Note, from the project list, that the fourth most profitable project (project2)according to the index is not chosen while two projects with lower index values areincluded.

We may try to change the budget restriction and study how the reward is increasingand the optimal set of projects is changing. In our example, the optimal reward is constant,243, for budget restriction in the interval [99, 103). It is 248 for budgets in [103, 109).Considering risks and cost of capital, it may not be worth an additional investment of103 − 99 = 4 to increase expected rewards by 248 − 243 = 5. However, increasing the

Page 439: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

426 Pharmaceutical Statistics Using SAS: A Practical Guide

budget to 109 increases the reward to 258 and, with a budget of 112, a reward of 278 ispossible. The last option corresponds to running the five projects with highest index.

Although the Pearson Index is useful, it is not perfect. Its adequacy depends on theextent to which it captures the options facing a drug developer as regards projects ingeneral. The index captures the obvious option on any project as regards costs: that is tosay, to abandon projects that no longer have any chance of being successful. Other optionsthat it does not capture, however, include the option to revise decisions in the light ofdeveloping market information and the option to revise investment decisions in the light ofchanges in the rate of interest (Senn, 1998; Senn and Rosati, 2002). For a generalintroduction to this field, see Dixit and Pindyck (1994).

14.9 SummaryDecision analysis is a general approach to decisions, based on problem structuring,quantifying, and optimizing. In principle, it can be applied to any decision. We have seen anumber of examples from drug development, with emphasis on clinical programs and trials.The examples range from the design of a single trial (sample size, sequential design), overthe question about optimal dose, to the design of a program and project prioritization. Thelist could of course be expanded. In particular, more examples could be taken fromdiscovery and commercial perspectives. An attractive feature of decision analysis is that itoften promotes cross-skill cooperation and is a useful tool to facilitate mutualunderstanding of each others’ ideas.

It must be stressed, however, that decision analysis must be handled with care. Within adecision model, it is straightforward to optimize the decision. However, this decision maybe quite bad if the decision model is not a good enough model for the real problem. Thereis a clear risk of getting a suboptimal decision as a result of a model that is too simplified.On the other hand, making models too complicated gives a similar risk. It is then hard tosee through the assumptions and to assess their validity. The inherently quantitative natureof a decision analysis may constitute a temptation to overvalue aspects that are easilymeasured and ignore qualitative aspects. It is important to realize that ethics must getpriority over profit, that a project cannot always be analyzed separately from long-termcompany strategy, that psychology sometimes is more important than cold rationalism.The conclusion is that decision analysis by all means should be applied in pharmaceuticaldevelopment but that the applications should be made with care.

AcknowledgmentsWe thank Axel Nilsson and Magnus Kjaer for significant improvements of the SAS code.

ReferencesAnscombe, F.J. (1963). “Sequential medical trials.” Journal of the American Statistical Association.

58, 365–383.Armitage, P. (1975). Sequential Medical Trials. Second edition. London: Blackwell Scientific

Publications.Bather, J. (2000). Decision Theory: an Introduction to Dynamic Programming and Sequential

Decisions. Chichester: Wiley.Berger, J.O. (1985). Statistical Decision Theory and Bayesian Analysis. Second edition. New York:

Springer-Verlag.Bergman, S.W., Gittins, J.C. (1985). Statistical Methods for Pharmaceutical Research Planning.

New York: Marcel Dekker.

Page 440: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Chapter 14 Decision Analysis in Drug Development 427

Berry, D.A., Muller, P., Grieve, A.P., Smith, M., Parke, T., Balazek, R., Mitchard, N., Krams, M.(2000). “Adaptive Bayesian Designs for Dose-Ranging Drug Trials.” In Case Studies in BayesianStatistics V. C. Gatsonis, R. E. Kass, B. Carlin, A. Carriquiry, A. Gelman, I. Verdinelli, and M.West, eds. New York: Springer-Verlag. 99–181.

Bertolli, J., Hu, D.J., Nieburg, P., Macalalad, A., Simonds, R.J. (2003). “Decision analysis to guidechoice of interventions to reduce mother-to-child transmission of HIV.” AIDS. 17, 2089–2098.

Brown, M.M., Brown, G.C., Sharma, S., Landy, J. (2003). “Health care economic analyses andvalue-based medicine.” Survey of Ophthalmology. 48, 204–223.

Burman, C.-F., Hamren, B., Olsson, P. (2005). “Modelling and simulation to improvedecision-making in clinical development.” Pharmaceutical Statistics. 4, 47–58.

Burman, C.-F., Senn, S. (2003). “Examples of option values in drug development.” PharmaceuticalStatistics. 2, 113–125.

Carlin, B.P., Kadane, J.B., Gelfand, A.E. (1998). “Approaches for optimal sequential decisionanalysis in clinical trials.” Biometrics. 54, 964–975.

Claxton, K., Posnett, J. (1996). “An economic approach to clinical trial design and researchpriority-setting.” Health Economics. 5, 513–524.

Committee for Proprietary Medicinal Products (CPMP) (2001). “Points to consider on applicationwith 1. Meta-analyses 2. One pivotal study.” London: European Evaluation Agency.

Cooper, N.J., Sutton, A.J., Abrams, K.R. (2002). “Decision analytic economic modelling within aBayesian framework: application to prophylactic antibiotics use for Caesarean section.”Statistical Methods in Medical Research. 11, 491–512.

Cornfield, J. (1969). “Sequential trials, sequential analysis and the likelihood principle.” TheAmerican Statistician. 20, 18–22.

Darken, P.F., Ho, S.-Y. (2004). “A note on sample size savings with the use of a singlewell-controlled clinical trial to support the efficacy of a new drug.” Pharmaceutical Statistics. 3,61–63.

DiMasi, J.A., Hansen, R.W., Grabowski, H.G. (2003). “The price of innovation: new estimates ofdrug development costs.” Journal of Health Economics. 22, 151–185.

Dixit, A.K., Pindyck, R.S. (1994). Investment under uncertainty. Princeton: Princeton UniversityPress.

Elwyn, G., Edwards, A., Eccles, M., Rovner, D. (2001). “Decision analysis in patient care.” TheLancet. 358, 571–574.

FDA (U.S. Department of Health and Human Services. Food and Drug Administration) (1998).“Guidance for industry: Providing clinical evidence of effectiveness for human drug andbiological products.” http://www.fda.gov/cder/guidance/1397fnl.pdf.

FDA (U.S. Department of Health and Human Services. Food and Drug Administration) (2004).“Challenge and Opportunity on the Critical Path to New Medical Products.”http://www.fda.gov/oc/initiatives/criticalpath/whitepaper.html.

FDA (U.S. Department of Health and Human Services. Food and Drug Administration) (2006).“Critical Path Opportunities List.”http://www.fda.gov/oc/initiatives/criticalpath/reports/opp list.pdf.

Fisher, L.D. (1999). “One large, well-designed, multicenter study as an alternative to the usual FDAparadigm.” Drug Information Journal. 33, 265–271.

Freedman, L.S., Spiegelhalter, D.J. (1989). “Comparison of Bayesian with group sequential methodsfor monitoring clinical trials.” Controlled Clinical Trials. 10, 357–367.

French, S (1989). Readings in Decision Analysis. London: Chapman and Hall.French, S., Rıos Insua, D. (2000). Statistical Decision Theory. London: Arnold.Gittins, J. and Pezeshk, H. (2000). “A behavioral Bayes method for determining the size of a

clinical trial.” Drug Information Journal. 34, 355–363.Graham, G., Gupta, S., Aarons, L. (2002). “Determination of an optimal regimen using a Bayesian

decision analysis of efficacy and adverse effect data.” Journal of Pharmacokinetics andPharmacodynamics. 29, 67–88.

Page 441: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

428 Pharmaceutical Statistics Using SAS: A Practical Guide

Howard, R.A., Matheson, J.E. eds. (1984). The Principles and Applications of Decision Analysis.Part I–II. Menlo Park, CA: Strategic Decision Group.

Kadane, J.B., Vlachos, P.K. (2002). “Hybrid methods for calculating optimal few-stage sequentialstrategies: data monitoring for a clinical trial.” Statistics and Computing. 12, 147–152.

Lehmann, E. L. (1986). Testing Statistical Hypothesis, New York: Wiley.Lindley, D. V. (1997). “The choice of sample size.” The Statistician. 46, 129–138.Liu, J.Y., Finlayson, S.R.G., Laycock, W.S., Rothstein, R.L., Trus, T.L., Pohl, H., and Birkmeyer,

J.D. (2003). “Determining an appropriate threshold for referral to surgery for gastroesophagealreflux disease.” Surgery. 133, 5–12.

O’Hagan, A., Stevens, J. (2001). “Bayesian assessment of sample size for clinical trials ofcost-effectiveness.” Medical Decision Making. 21, 219–230.

Pallay, A. (2000). “A decision analytic approach to determining sample sizes in a Phase IIIprogram.” Drug Information Journal. 34, 365–377.

Pallay, A., Berry, S.M. (1999). “A decision analysis for an end of Phase II go/stop decision.” DrugInformation Journal. 33, 821–833.

Parmigiani, G. (2002). Modeling in Medical Decision Making. New York: Wiley.Pearson, A.W. (1972). “The use of ranking formulae in R & D projects.” R & D Management. 2,

69–73.Pezeshk, H. (2003). “Bayesian techniques for sample size determination in clinical trials: a short

review.” Statistical Methods in Medical Research. 12, 489–504.Poland, B., Wada, R. (2001). “Combining drug-disease and economic modelling to inform drug

development decisions.” Drug Discovery Today. 6, 1165–1170.Protheroe, J., Fahey, T., Montgomery, A.A., Peters, T.J., Smeeth, L. (2000). “The impact of

patients’ preferences on the treatment of atrial fibrillation: observational study of patient baseddecision analysis.” British Medical Journal. 320, 1380–1384.

Raiffa, H. (1968). Decision Analysis—Introductory Lectures on Choices under Uncertainty,Reprinted 1997, New York: McGraw Hill.

Raiffa, H., Schlaifer, R. (1961). Applied Statistical Decision Theory. Reprinted 2000. New York:Wiley.

Rosenkranz, G. (2002). “Is it possible to claim efficacy if one of two trials is significant while theother just shows a trend?” Drug Information Journal. 36, 875–879.

Savage, L.J. (1954). The Foundations of Statistics. New York: Wiley.Senn, S.J. (1996). “Some statistical issues in project prioritization in the pharmaceutical industry.”

Statistics in Medicine. 15, 2669–2702.Senn, S.J. (1997). Statistical Issues in Drug Development. Chichester: Wiley.Senn, S.J. (1998). “Further statistical issues in project prioritization in the pharmaceutical

industry.” Drug Information Journal. 32, 253–259.Senn, S.J., Rosati, N. (2002). “Project selection, decision analysis and profitability in the

pharmaceutical industry.” Business Briefing: Pharmatech 2002. 18–21, London.http://www.wma.net/e/publications/pdf/2001/senn.pdf

Wald, A. (1950). Statistical Decision Functions. New York: Wiley.Whitehead, J. (1997). The Design and Analysis of Sequential Medical Trials. Second edition. New

York: Wiley.Yin, Y. (2002). “Sample size calculation for a proof of concept study.” Journal of Biopharmaceutical

Statistics. 12, 267–276.Zipfel, A. (2003). “Modeling the probability-cost-profitability architecture of portfolio management

in the pharmaceutical industry.” Drug Information Journal. 37, 185–205.

Page 442: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index

A

A-optimality 155 AB option, NPAR1WAY procedure 126 acceptance limit 71

See also validation See also validation of analytical methods

accepted reference value 94 accidental bias 214 accuracy of analytical procedures 85–88 AdaBoost algorithm 11–12

See also boosting model building 27–33 using IML_SPLIT module 17–21

%AdaBoost macro 17–21 ADAPT II program 158 adaptive quadrature 327 adaptive resampling and combining (ARCing)

14 ADD statement, REG procedure 58–59 allocation in randomized clinical trials 213–233

accidental bias 214 balancing across centers 224–227 baseline covariates 228–233, 261–262 constrained block randomization 220–224 covariate-adaptive allocation 228–229 imbalance in 213–214 minimization algorithms 229–233 permuted block randomization 214–219 treatment group splitting 221–224

analysis of incomplete data 314–356 categorical data 322–340 data setting and modeling framework 318 death, data missing because of 109–110 missing at random (MAR) 320–322 missing completely at random (MCAR) 313,

314, 319, 343–347 missing not at random (MNAR) 340–347 sensitivity analysis 347–356

analytic method validation 69–94 accuracy and precision 85–88 criteria for 73–74 decision rule 88–92 limits of 92–93 linearity 83–85 objectives of analytical methods 71 range of analytic procedures 92–93

repeatability 85 response function (calibration curve) 74–83 terminology 94

analytical procedures, error of 85–88 ANOVA model

one-factor with repeated measures 106–113 two-factor, power evaluation in 102–106

Ansari-Bradley test 125–126 NPAR1WAY procedure 126–127

ARCing (adaptive resampling and combining) 14

ARM= option, FACTORS statement (PLAN) 221–223

assays See analytic method validation

Assessment node (SAS Enterprise Miner) 66 %ASSIGN macro 230–233 asthma trial 283–284 asymmetry factor (5-parameter logistic model)

80 AUC (plasma concentration curve) 198

See also pharmacokinetic data analysis dose proportionality 204–207

available case methods 320

B

β-expectation tolerance intervals 89–91 back-calculated quantities 82–83 backward elimination procedures 52 backward induction 393, 407 backward step (optimal design computation)

158 balanced-across-centers allocation 224–227 Bartholomew trend test, sample size calculations

for 288 baseline covariates, allocations based on

228–233, 261–262 Bayes criterion 393 Bayesian sample size calculations 403–405 Behrens-Fisher problems 119 β-expectation tolerance intervals 89–91 Beta regression models 172–176

information matrix 173 with logic link 176–181

bias in random subject allocation 213–214 bias-variance trade-off 12 bin widths 48 binary models 159–165

Page 443: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

430 Index

BINOMIAL option, EXACT statement (FREQ) 244

bioanalytical method validation See analytic method validation

bioassay studies 152 bioequivalence testing 197–204

replicate cross-over designs 201–204 bivariate binary Cox model 181 bivariate probit models 181–184 BLOCK= option, FACTORS statement (PLAN)

216–217, 221–222 block randomization in parallel design studies

100–101 block size

balancing across centers 224–227 constrained block designs 220–221 permuted block designs 215, 217–219

body weight data 99 Bonferroni-adjusted t-test 113 %Boost macro 22–25

model building 27–33 boosting 10–27

evolution of 11 generalized boosting algorithm 12, 13,

22–26 implementing in SAS 14–26 misclassification rates and 15–16, 26, 31 model building with 27–33 Naive-Bayes 14 neural network 13–14 properties of 26–27 weak learning algorithms 11–14 with recursive partitioning 14–16 with unequally weighted observations 16–17

%BOOTSTRAP macro 147–148 bootstrap method (prediction error) 47 bounded-response models (Beta models)

172–176 with logic link 177–181

BOUNDS statement, NLMIXED procedure 80 Bradley-Blackwood test 373–376 BY statement, MIXED procedure 76

C

c-optimality 155 calibration curve (response function) 74–83

back-calculated quantities and inverse predictions 82–83

fitting models 74 linear and polynomial models 75–76 nonlinear models 76–79

precision profiles 79–81 calibration samples (pre-study validation) 72,

94 canonical covariates analysis (CCA) 33–36 canonical discriminant analysis (CDA) 36 categorical data, missing 322–340

marginal models 322–324, 327, 329–335 random-effects models 324–330, 335–340

CC (complete case) analysis 314, 320 CCA (canonical covariates analysis) 33–36 CCC (concordance correlation coefficient) 363

inter-rater reliability 369–370 test-retest reliability 372–376

CDA (canonical discriminant analysis) 36 central randomization algorithms 225 change sensitivity (responsiveness) 376, 378 CLASS statement

GLIMMIX procedure 333 NPAR1WAY procedure 121

classical error rates in sample-size analysis 238, 239, 249–250

classification methods for drug discovery 7–42 boosting 10–27 model building with boosting 27–33 partial least squares for discrimination 33–

42 clearance, time stationarity of 207–209 clinical dose-ranging studies

See dose-response studies clinical pharmacokinetic studies 153

See also pharmacokinetic data analysis See also pharmacokinetic models with

multiple per-patient measurements clinical pharmacology research, exploratory

199–201 clinical testing 3–4 clinical trial allocation (randomized) 213–233

balancing across centers 224–227 baseline covariates 228–233, 261–262 constrained block randomization 220–224 permuted block randomization 214–219

clinical trials incomplete data in 314–316 longitudinal 313, 314 optimal number of 400–403 Phase III trials 4, 389 sequential designs in 406–412

closed family of hypotheses 297 closed testing on multiple contrasts 297–298,

305 CMERGE variable (%OptimalDesign1) 162

Page 444: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 431

coefficient-of-variation (CV) 79 complete block design, randomized 100–101 complete case (CC) analysis 314, 320 complete randomization 214 computation models, building

See model building concordance correlation coefficient (CCC) 363

inter-rater reliability 369–370 test-retest reliability 372–376

conditional independence models 325 confidence intervals 307-309 confirmatory bioequivalence assessment 199 CONST1, CONST2 variables

(%OptimalDesign1) 162 constrained block randomization 220–224 content validity 376 continuous logistic models 153, 165–172

with unknown parameters in variance 169–172

CONTRAST statement, MIXED procedure detection of dose-response trends 283–284 monotonic dose-response relationships

112–113 contrast tests in dose-response studies 281–282

asthma trial (example) 283–284 diabetes trial (example) 282–283 linear tests 281, 296–297, 304 maximin tests 282 modified linear tests 282 multiple-contrast tests vs. 295 sample size calculations 287–289

control, choosing for dose-response studies 278–279

CONVC variable (%OptimalDesign1) 161 convergent validity 377 CORR procedure 371 cost-based designs 190–192 cost constraints, models with 190–192 covariate-adaptive allocation 228–229

See also minimization algorithms criteria for validation 73–74 Cronbach’s alpha 370–372 cross-over dose-response studies 278 cross-validation 47

See also prediction error generalized boosting and 22

crucial Type I and II error rates 238, 239, 249–253, 260–263

%CrucialRates macro 252, 265–271 CV (coefficient-of-variation) 79

D

D-optimality 155, 156 bivariate probit models 183–184 bounded-response models with logic link

177–181 four-parameter logistic model with

continuous response 167–169 logistic model with power variance model

171–172 pharmacokinetic models 187–190, 191–192 two-parameter logistic models 160–161,

164–165 data, missing

See analysis of incomplete data data analysis in toxicology studies 99 Data Partition node (SAS Enterprise Miner) 64 Data Set Attributes node (SAS Enterprise Miner)

65 data transformations 129 death, data missing because of 109–110 decision analysis 385–426

optimal dose selection 412–421 optimal sample size 397–406 project prioritization 421–426 sequential designs in clinical trials 406–412 structure of 392–394

decision rule 88–92 decision trees 386–392, 394–396

evaluating 389–392 definite quantitative assays 70

See also validation of analytical methods depression trial 317–318

See also analysis of incomplete data design optimization

See optimal experimental designs design region for optimal designs 157–158 diabetes trial 282–283 Diggle-Kenward model 340–347

sensitivity analysis 348–350, 352–356 direct likelihood analyses 320–321 discovery data classification 7–42

boosting 10–27 model building with boosting 27–33 partial least squares for discrimination 33–

42 DISCRIM procedure 38–41 discriminant analysis 59–60 discriminant validity 377–378

Page 445: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

432 Index

discrimination, partial least squares (PLS) for 33–42

definition and theorem 34 implementing PLS-LDA in SAS 38–41

dispersion of two distributions, comparing 125–129

See also two-sample experiments distribution-free methods

See nonparametric methods divergent validity 377 dose-escalation studies 277 dose linearity assessment 204–209 dose proportionality 204–207 dose-response studies 152, 273–310

See also MED estimation See also stepwise procedures in dose-finding

studies choosing hypothesis for 278–279 contrast tests 281–284, 287–289, 295 cross-over studies 278 design considerations 277–280 factorial 278 Helmert contrasts 296 isotonic tests 284–286 monotonic dose-response relationships

112–113 Monte Carlo method 302–303 noninferiority testing 279–280 nonmonotone response curves 304–306 nonmonotonic relationships 112–113 one-sided 280 partitioning principle 305–309 quantal dose-response models 159–165 regression modeling 289–294 reverse Helmert contrasts 296, 302–303,

304–305 trend detection 280–289

dose selection 412–421 from limited data 415–421 models without uncertainty 413–415

dropouts 314, 318–319 See also analysis of incomplete data

drug development statistics 1–6 drug discovery classification methods 7–42

boosting 10–27 model building with boosting 27–33 partial least squares for discrimination

33–42 drug toxicity studies

See nonclinical safety assessment

DSCF (Dwass-Steel-Critchlow-Fligner) procedure 136–139, 143–144

Dunn’s procedure vs. 139, 144 DTREE procedure 386–387, 391–392, 396

MODIFY statement 396, 422–423 PROBIN option 392 project prioritization 422–423

%DUNN macro 135–136 Dunnett’s t-test 113 Dunn’s procedure 135–136, 140, 143–144

DSCF procedure vs. 139, 144 Miller’s method vs. 142–144

Dwass-Steel-Critchlow-Fligner (DSCF) procedure 136–139, 143–144

Dunn’s procedure vs. 139, 144

E

E-optimality 155 effective dose, minimum

See MED estimation effective exposure levels 293 EM (Expectations-Maximization) algorithm

321–322 Emax models

See continuous logistic models EMPIRICAL option, %GLIMMIX macro 324,

330 ensemble techniques 8, 14 equivalence theorem 157 error, Type I

See Type I error rates error, Type II

See Type II error rates error of analytical procedures 85–88 ERROR statement, %GLIMMIX macro 330 ESTIMATE statement, MIXED procedure 105

monotonic dose-response relationships 112–113

pharmacokinetic data analysis 208 estimating minimum effective dose

See MED estimation exact Kruskal-Wallis test 133–134 EXACT statement, FREQ procedure 286

BINOMIAL option 244 EXACT statement, NPAR1WAY procedure

133–134 WILCOXON option 133

exercise trial 316 See also analysis of incomplete data

Expectations-Maximization (EM) algorithm 321–322

Page 446: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 433

experimental designs, optimal See optimal experimental designs

exploratory clinical pharmacology research 199–201

exposure levels 293

F

F test 281 factor analysis 376, 379–382 FACTOR procedure 380

NFACT option 380 REORDER option 380 ROTATE option 380 SCREE option 380 SIMPLE 380

factorial dose-response studies 278 factorial stratification 227 FACTORS statement, PLAN procedure

101–102, 216–217, 221–222 ARM= option 221–223 BLOCK= option 216–217, 221–222

familywise error rate 298 5-parameter logistic regression models 79–80 fixed-sequence estimation for minimum

effective dose 298, 301–302 Fligner-Policello test 127–129

rank-transformations and 129 forward selection procedures 52, 55–56 forward step (optimal design computation) 158 4-parameter logistic regression models 76–78,

80–81 back-calculated quantities and inverse

predictions 82–83 linearity assessment 83–84

four-parameter logistic models See continuous logistic models

FPARA module 188 %FPSTATISTIC macro 128–129 FREQ procedure 244

EXACT statement 244, 286 Jonckheere test 286–287 Kappa statistic calculations 365 TABLES statement 286

G

GAM procedure 60–61 Y= option 60

GAUSS value, METHOD= option (NLIN) 77 GEE (generalized estimating equations)

322–323 weighted (WGEE) 322–323, 329

general optimal design problem 152–159 generalized additive regression 60–61 generalized boosting algorithm 12

comparison of 13 implementing 22–26

generalized estimating equations (GEE) 322–323

weighted (WGEE) 322–323, 329 generalized linear mixed models (GLMMs)

325–329 fitting 338

Gentle AdaBoost algorithm 12–13, 22–26 model building 27–33

Gini Index 15–16 See also boosting

%GLIMMIX macro 325 EMPIRICAL option 324, 330 ERROR statement 330 LINK statement 330 marginal model (example) 330–332 METHOD= option 333 PARMS statement 339 random-effects model (example) 338–340 REPEATED statement 330, 334 STMTS= statement 330, 338

GLIMMIX procedure 325 CLASS statement 333 marginal model (example) 332–335 MODEL statement 333–334 random-effects model (example) 335–338 RANDOM statement 292, 334, 336

GLMMs (generalized linear mixed models) 325–329

fitting 338 GLMPOWER procedure 257, 262 global influence approach to sensitivity analysis

354–356 go/no go decisions 386–392, 394–397 GRID parameter (%OptimalDesign1) 161 GROUPWEIGHTS statement, POWER

procedure 244–245 Gumbel model 181

H

Helmert contrasts in dose finding 296 reverse Helmert contrasts 296, 302–303,

304–305 Hill models

See continuous logistic models HOLD option, PARMS statement

(%GLIMMIX) 339

Page 447: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

434 Index

Hsu-Berger method 307 hypotheses

choosing for dose-response studies 278–279 closed family of 297

I

I-optimality 156 ICC (intraclass correlation coefficient) 363

inter-rater reliability 369–370 test-retest reliability 372–376

ignorability 319 See also analysis of incomplete data

imbalance in subject allocation 213–214 See also allocation in randomized clinical

trials IML_SPLIT module 17–21 incomplete data, analysis of

See analysis of incomplete data influential points in training sets 48 information matrix 154

Beta regression models 173 bivariate probit models 182 bounded-response models with logic link

179, 181 normalized by cost 190–192 regression model with unknown parameters

in variance 170 two-parameter logistic models 162

Input Data Source node (SAS Enterprise Miner) 64

inter-rater reliability 362–363 multiple raters 367–369 rating scale data 369–370

intermediate precision 85–88 intermittent missingness 319 internal consistency of outcome measures 362,

370–372 intra-rated reliability 362 intraclass correlation coefficient

See ICC inverse response functions 82–83 IRLS (iteratively reweighted least squares) 324 isotonic tests in dose-response studies 284–286 iteratively reweighted least squares (IRLS) 324

J

JMP software 158 Jonckheere test 286–287 JT option, TABLES statement (FREQ) 286

K

Kappa statistic 363–365 inter-rater reliability 367–369 multiple rates 367–369 proportion of positive (negative) agreement

365–367 Kruskal-Wallis test 131–134

large-sample 133, 134 nonparametric power analysis 147–149 NPAR1WAY procedure 132–134 power calculations in k-sample case 146

%KWSS macro 147–149

L

large-sample Kruskal-Wallis test 133, 134 Lasso shrinkage method 52 last observation carried forward (LOCF) 313,

314, 319–320 Latin square design studies 99

randomization in 101–102 %LATINSQ macro 101–102 LDA (linear determinant analysis) 8, 33–38

See also partial least squares for discrimination

implementing PLS-LDA in SAS 38–41 leaps and bounds (subset algorithms) 52 leverage value 62–63 likelihood-based MAR analysis 320–321 limit of detection (LOD) 93 limits of quantification 92–93 linear classification methods 8 linear contrast tests 281, 296–297

nonmonotone dose-response curves 304 linear determinant analysis

See LDA linear regression models 58–59, 75–76

calibration curve (response function) 75–76 in dose-response studies 289–291 multiple linear regression 58–59

linearity of analytical procedures 83–84 linearization-based marginal models 323–324 LINK statement, %GLIMMIX macro 330 LLOQ (lower limit of quantification) 92 local influence approach to sensitivity analysis

350–356 global influence vs. 354–356

locally optimal designs 156–157 LOCF (last observation carried forward) 313,

314, 319–320

Page 448: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 435

LOD (limit of detection) 93 LOESS value, Y= option (GAM) 60 logarithmic data transformations 129 LOGISTIC procedure for optimal dose selection

417–419 logistic regression models 8, 59, 76–78

back-calculated quantities and inverse predictions 82–83

continuous models 153, 165–172 five-parameter 79–80 four-parameter 76–78, 80–84 linearity assessment 83–84 optimal design, continuous response

165–169 optimal design, quantal models 159–165 optimal design, unknown parameters in

variance 169–172 precision profiles 80–81 two-parameter 160–161, 164–165 with power variance model 171–172

logit link models 176–181 logit models, quantal 159–165 LogitBoost algorithm 12–13, 22–24

model building 27–33 longitudinal clinical trials 313, 314

See also analysis of incomplete data lower limit of quantification (LLOQ) 92 lower-tailed version of Wilcoxon Rank Sum test

122 LP procedure 425

M

MAR (missing at random) modeling 314–316, 329

analysis of incomplete data 320–322 assumption of MAR 342 direct likelihood analyses 320–321 Expectations-Maximization (EM) algorithm

321–322 MNAR modeling vs. 343–347 multiple imputation methods 321

marginal models for missing categorical data 322–324, 327

%GLIMMIX macro for (example) 330–332 GLIMMIX procedure for (example)

332–335 linearization-based 323–324 random-effects models vs. 329–330

marginal quasi-likelihood (MQL) estimates 327

MARQUARDT value, METHOD= option (NLIN) 77

mastitis data example 316–317 See also analysis of incomplete data

maximal procedure 221 maximin contrast tests 282 MAXIMIT variable (%OptimalDesign1) 162 maximum likelihood estimates (MLEs) 155 MCAR (missing completely at random)

assumption 313, 314, 319 MNAR modeling vs. 343–347

MCID (minimal clinically important differences) 376, 382

MEANS procedure 128 measurement error, analytical procedures 85–88

profiles of 89–92 MED (minimum effective dose) estimation 274,

294–309 closed testing on multiple contrasts 297 estimation in general case 305–306 estimation under monotonicity assumption

294–295, 303–305 fixed-sequence estimation 298, 301–302 multiple-contrast tests 295–297, 299,

302–303 partitioning principle 305–306 shortcut testing procedure 306–307 simultaneous confidence intervals 307–309 step-down estimation 298, 299–301

merging constant 162 METHOD= option

GLIMMIX procedure 333 NLIN procedure 77 NLMIXED procedure 78

MI (multiple imputation) methods 321 Miller’s method for all group comparison

140–144 minimal clinically important differences (MCID)

376, 382 minimally effective exposure level 293 minimax optimality 156, 393 minimization algorithms (for allocation)

229–233 minimum effective dose (MED) estimation

See MED estimation misclassification rates, and boosting 15–16, 26,

31 missing at random (MAR) modeling

See MAR modeling

Page 449: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

436 Index

missing categorical data See categorical data, missing

missing completely at random (MCAR) 313, 314, 319

MNAR modeling vs. 343–347 missing data

See analysis of incomplete data missing not at random

See MNAR missingness

intermittent 319 monotone 318–319 nonmonotone 319

mixed-effects nonlinear regression models 186–190

MIXED procedure See also REPEATED statement, MIXED

procedure BY statement 76 CONTRAST statement 112–113, 283–284 ESTIMATE statement 105, 112–113, 208 monotonic dose-response relationships

112–113 pharmacokinetic data analysis 198, 200–208 RANDOM statement 111, 198, 201, 207 regression modeling for dose-response

studies 290 SOLUTION option 75 trends in dose-response relationship,

detecting 283–284 TYPE= option 203 WEIGHT statement 75

MIXPRINTALL option, STMTS= statement (%GLIMMIX) 330

MLEs (maximum likelihood estimates) 155 MNAR (missing not at random) 314–316,

340–347 analysis of incomplete data 340–345 MAR modeling vs. 343–347 MCAR assumption vs. 343–347 sensitivity analysis 348–350, 352–356

model building 45–67 AdaBoost algorithm 11–12 %Boost macro 22–25 Gentle AdaBoost algorithm 27–33 LogitBoost algorithm 27–33 SAS Enterprise Miner for 63–67 statistical procedures for 58–61 training and test sets 46–51, 61–63 variable selection 51–58

when observations not in training sets 61–63 with boosting 27–33

model prediction error 46, 47 MODEL statement, GLIMMIX procedure

333–334 OUTPREDM option 75, 290 SOLUTION option 334

modified linear contrast tests 282 MODIFY statement, DTREE procedure 396,

422–423 monotone missingness 318–319 monotonic dose-response relationships 112–113 monotonicity assumption, MED estimation

under 294–295, 303–305 Monte Carlo method for dose finding 302–303 mortality rates 240 MQL (marginal quasi-likelihood) estimates 327 multi-parameter continuous logistic models

See continuous logistic models multiple-contrast tests for MED estimation

295–297, 299, 302–303 multiple imputation (MI) methods 321 multiple linear regression 58–59 multivariate probit model 182

N

Naive-Bayes boosting 14 negative agreement, proportion of 365–367 neural network boosting 13–14 NEWRAP value, TECHNIQUE= option

(NLMIXED) 78 NFACT option, FACTOR procedure 380 NIPALS algorithm 34 NLIN procedure

fitting nonlinear models 76–78 METHOD= option 77 optimal dose selection 417–419 PARAMETERS statement 77 _WEIGHT_ statement 77

NLMIXED procedure 77, 78–79 BOUNDS statement 80 conditional independence models 325 fitting GLMM 338 fitting nonlinear regression models 78–79 METHOD= option 78 sigmoid regression models for dose-response

curves 292 TECHNIQUE= option 78–79

NLP procedure 400 optimal dose selection 414–415

Page 450: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 437

nonclinical safety assessment 97–115 data analysis 99 key statistical aspects 98 one-factor design with repeated measures

106–113 power evaluation in two-factor model

102–106 randomization 99–102

nonclinical statistical support 2–3 noninferiority testing (dose-response studies)

279–280 nonlinear models, optimal experimental designs

for See optimal experimental designs

nonlinear regression models 164–165 calibration curve (response function) 76–79 fitting with NLIN procedure 76–78 fitting with NLMIXED procedure 78–79 in dose-response studies 289, 291–294 mixed-effects with multiple responses

186–190 optimal design 165–169 optimal design, unknown parameters in

variance 169–172 partially nonlinear models 167

nonmonotone dose-response curves 304–306 nonmonotone missingness 319 nonmonotonic dose-response relationships

112–113 nonparametric methods 117–149

Kruskal-Wallis test 147–149 one-way layout 135–144 parametric approaches vs. 147 power determination 144–149 two independent samples 118–129

nonparametric trend tests 286 nonrandom missing data 314–316, 340–347

sensitivity analysis 348–350, 352–356 nonresponse process

See MCAR assumption %NOPT macro 400 normal bin width 48 NPAR1WAY procedure 121, 124–125

AB option 126 Ansari-Bradley test 126–127 CLASS statement 121 EXACT statement 133–134 Kruskal-Wallis test 132–134 ODS statement 121 VAR statement 121 WILCOXON option 121, 133

%NPARMCC macro 140–144 NRRIDG value, TECHNIQUE= option

(NLMIXED) 78

O

objective of analytical methods 71 observer bias 213–214 ODS GRAPHICS statement 61 ODS statement, NPAR1WAY procedure 121 one-factor ANOVA design with repeated

measures 106–113 one-sided dose-response studies 280 one-way layout 129–144

Kruskal-Wallis test 131–134, 146–149 nonparametric multiple comparison

procedures 135–144 %ONEWEEK macro 108 OPTEX procedure 158 optimal dose selection 412–421

from limited data 415–421 LOGISTIC procedure 417–419 models without uncertainty 413–415 NLIN procedure 417–419 NLP procedure 414–415

optimal experimental designs 151–193 backward step 158 Beta regression models 172–176 bivariate probit models 181–184 bounded response (logit link) 177–181 design region 157–158 forward step 158 general optimal design problem 152–159 models with cost constraints 190–192 nonlinear regression models with continuous

response 165–169 partially nonlinear models 167 pharmacokinetic models with multiple per-

patient measurements 184–190 quantal dose-response models 159–165 regression models with unknown variance

function parameters 169–172 software for generating 158

optimal sample size in decision analysis 397–406

%OptimalDesign1 macro 161, 167 CMERGE variable 162 CONST1, CONST2 variables 162 CONVC variable 161 GRID parameter 161 MAXIMIT variable 162 PARAMETER parameter 161

Page 451: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

438 Index

%OptimalDesign1 macro (continued) POINTS parameter 161 WEIGHTS parameter 161

%OptimalDesign2 macro 177 %OptimalDesign3 macro 179–181 %OptimalDesign4 macro 183–184 optimality

See also D-optimality A-optimality 155 c-optimality 155 criteria 155–156 E-optimality 155 I-optimality 156 minimax optimality 156, 393 semi-Bayesian approach 393

OPTIONS= option, STMTS= statement (%GLIMMIX) 330

ORDERVARS module 53–55 outcome measures 361–383 outcome measures, reliability 361–376

inter-rater reliability 362–363, 367–370 test-retest reliability 362, 372–376

outcome measures, validity 361, 376–382 convergent and divergent 377 discriminant 377–378 responsiveness 376, 378

outliers in training sets 48 OUTPREDM option, MODEL statement

(GLIMMIX) 75, 290 OUTPUT statement, PLAN procedure 101–102

RELRISK option 244

P

p-values 241 pairwise comparisons, one-way layout 135–144 pairwise contrasts in dose finding 296, 299 parallel design studies 99, 278

randomization in 100–101 PARAM value, Y= option (GAM) 60 PARAMETER parameter (%OptimalDesign1)

161 PARAMETERS statement, NLIN procedure 77 PARMS statement, %GLIMMIX macro 339

HOLD option 339 partial least squares (PLS) for discrimination

33–42 definition and theorem 34 implementing PLS-LDA in SAS 38–41

partial least squares (PLS) for variable selection 51–52

partially nonlinear models 167

partitioning principle to dose finding 305–306 simultaneous confidence intervals 307–309

pattern-mixture models (PMMs) 314, 319 PCA (principal components analysis) 8, 33–34 PCR (principal component regression) 51 Pearson Index 421–424 Pearson’s correlation coefficient 372–373 penalized quasi-likelihood (PQL) estimates

326–327 permuted block randomization 214–217

stratified 228 with variable block size 217–219

pharmacokinetic data analysis 197–209 bioequivalence testing 199–204 dose linearity assessment 204–209 MIXED procedure 198, 200–208 steady state 204, 207–208

pharmacokinetic models with multiple per-patient measurements

optimal design 184–190 regression models 186–187 two-component models 184–186 with cost constraints (example) 191–192 without cost constraints (example) 187–190

Phase III trials 4, 389 PK

See pharmacokinetic data analysis See pharmacokinetic models with multiple

per-patient measurements placements (Fligner-Policello test) 127 PLAN procedure

balanced-across-centers allocation 226–227 FACTORS statement 101–102, 216–217,

221–222 generating constrained randomization

schedules 221–224 generating Latin square design 101–102 generating permuted block schedules

216–217 OUTPUT statement 101–102 SEED= option 216 TREATMENTS statement 101–102, 226

plasma concentration curve (AUC) 198 See also pharmacokinetic data analysis dose proportionality 204–207

PLS (partial least squares) for discrimination 33–42

definition and theorem 34 implementing PLS-LDA in SAS 38–41

PLS (partial least squares) for variable selection 51–52

Page 452: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 439

PLS procedure 34, 38–41 PMMs (pattern-mixture models) 314 Pocock-Simon minimization algorithm 229–230 point-in-time analyses 378 points of influence in training sets 48 POINTS parameter (%OptimalDesign1) 161 polynomial regression models 76 population-averaged models 323–324

See also marginal models for missing categorical data

positive agreement, proportion of 365–367 power analysis

See sample-size analysis power evaluation (toxicology studies) 99,

102–106 POWER procedure 244–245, 246

GROUPWEIGHTS statement 244–245 SIDES= option 245

power variance model 171–172 PQL (penalized quasi-likelihood) estimates

326–327 PQL (pseudo-quasi-likelihood) estimates

326-327 pre-study validation

See validation See validation of analytical methods

precision of analytical procedures 85–88 precision profiles 79–81 %Predict macro 25–26 prediction error 46, 47 %PRF1FRM macro 113 principal component regression (PCR) 51 principal components analysis (PCA) 8, 33–34 PROBIN option, DTREE procedure 392 probit models, quantal (binary) 159–160,

181–184 PROBNORM function 128 project prioritization 421–426 proportion of positive (negative) agreement

365–367 proportionality, dose 204–207 pseudo-quasi-likelihood (PQL) estimates

326–327 psychometric properties of rating scales

See outcome measures

Q

QCA mortality rates (example) 240 quadratic regression models 76

quadrature rules for random-effects models 327–328

qualitative methods 70 QUANEW value, TECHNIQUE= option

(NLMIXED) 78 quantal dose-response models 159–165 quantification, limits of 92–93 quantitative assays, definite 70

See also validation of analytical methods quantitative methods 70–71 quasi-quantitative assays 71 questionnaire subscales, identifying 379–382

R

random-effects models for missing categorical data 324–330

%GLIMMIX macro for (example) 338–340 GLIMMIX procedure for (example)

335–338 marginal models vs. 329–330 quadrature rules for 327–328

RANDOM statement, GLIMMIX procedure 336

SUBJECT option 292, 336 TYPE= option 334, 336

RANDOM statement, MIXED procedure 111 pharmacokinetic data analysis 198, 201, 207

randomization (toxicology studies) 98–102 complete block design 100–101

randomized clinical trials, allocation in 213–233

balancing across centers 224–227 baseline covariates 228–233, 261–262 complete randomization 214 constrained block randomization 220–224 permuted block randomization 214–219

range of analytic procedures 92–93 RANK procedure 100–101 rank-transformations 129 RANTBL function 218 RANUNI function 100, 218 rating scales, psychometric properties

See outcome measures RCORR option, REPEATED statement

(MIXED) 331 Real AdaBoost

See AdaBoost algorithm receiver operator characteristic (ROC) curves

378 recursive partition models 8

Page 453: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

440 Index

recursive partitioning 12 boosting with 14–16

REG procedure 58–59 ADD statement 58–59 for forward selection 55–56 VAR statement 58–59

regression, ridge 52 regression models, Beta 172–176

with logic link 176–181 regression models, generalized additive 60–61 regression models, linear and polynomial

58–59, 75–76 in dose-response studies 289–291 quadratic models 76

regression models, logistic See logistic regression models

regression models, nonlinear See nonlinear regression models

regression models, principal component (PCR) 51

regression models, sigmoid 291–294 Regression node (SAS Enterprise Miner) 66 REGSPLIT.IML module 22 relative quantitative assays 71

See also validation of analytical methods reliability of outcome measures 361–376

inter-rater reliability 362–363, 367–370 test-retest reliability 362, 372–376

RELRISK option, OUTPUT statement (PLAN) 244

REML (restricted maximum likelihood modeling) 198

REORDER option, FACTOR procedure 380 repeatability of analytical procedures 85 repeated measures ANOVA 106–113 REPEATED statement, %GLIMMIX macro

334 TYPE= option 330

REPEATED statement, MIXED procedure 111 detection of dose-response trends 112–113 pharmacokinetic data analysis 198, 201 RCORR option 331 regression modeling for dose-response

studies 290 replicate cross-over design 201–204 %REPMEAS49 macro 112 reproducibility of analytical procedures 85 research hypothesis, choosing for dose-response

studies 278–279

response function (calibration curve) 74–83 back-calculated quantities and inverse

predictions 82–83 fitting models 74 linear and polynomial models 75–76 nonlinear models 76–79 precision profiles 79–81

responsiveness 376, 378 restricted maximum likelihood modeling

(REML) 198 reverse Helmert contrasts in dose finding 296,

302–303 non-monotone dose-response curves

304–305 ridge regression 52 robust bin width 48 robustness of decision analysis conclusions 396 ROC (receiver operator characteristic) curves

378 ROTATE option, FACTOR procedure 380

S

safety assessment, nonclinical See nonclinical safety assessment

sample-size analysis 237–264 classical (examples of) 243–248, 257–260 classical error rates 238, 239, 249–250 crucial Type I and II error rates 238, 239,

249–253, 260–263 decision analytic approaches 397–406 linear contrast tests and 287–289 macro code for automatic programming

265–271 statistical considerations summary 264–265

SAS Enterprise Miner Assessment node 66 boosting implementation 14 Data Partition node 64 Data Set Attributes node 65 Input Data Source node 64 model building 63–67 Regression node 66 Variable Selection node 64–65 with solubility data 63–67

SCREE option, FACTOR procedure 380 SEED= option, PLAN procedure 216 selection bias 213 selectivity of analytical procedure 74 semi-Bayesian optimality approach 393

Page 454: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 441

sensitivity analysis for incomplete data 347–356 global influence approach 354–356 local influence approach 350–356 MNAR 348–350, 352–356

sensitivity functions 157 Beta regression models 173 bounded-response models with logic link

181 regression model with unknown parameters

in variance 170–171 two-parameter logistic models 162–164

sensitivity to change (responsiveness) 376, 378 sequential decision problems 393 sequential designs in clinical trials 406–412 sequential testing method 103–104, 106–113 shift in location 119 shrinkage methods 52 SIDES= option, POWER procedure 245 sigmoid regression models for dose-response

curves 291–294 SIMPLE option, FACTOR procedure 380 %SimTests macro 302–303 simulation of optimal sample size 407–409 %SIMULQT macro 104–105 simultaneous confidence intervals 307-309 single-contrast tests

See contrast tests in dose-response studies smallest clinically meaningful difference 288 software for generating optimal designs 158 solubility data (example) 46–47

SAS Enterprise Miner with 63–67 training and test sets 49–51 variable ordering in 56–58

SOLUTION option MIXED procedure 75 MODEL statement (GLIMMIX) 334

spatial power covariance structure 111–112 specificity of analytical procedure 74 SPLINE value, Y= option (GAM) 60 SPLINE2 value, Y= option (GAM) 60 %Split macro 17 statistical decision theory

See decision analysis statistical procedures for modeling 58–61 statisticians, roles of 2–6 statistics in drug development 1–6 steady state (pharmacokinetics) 204, 207–208 step-down Dunnett test 299–301 step-down estimation for minimum effective

dose 298, 299–301

step-down procedure 298 stepwise procedures in dose-finding studies 52,

297–301, 302–303 fixed-sequence pairwise test 298, 301–302 pairwise multiple-contrast test 296, 299 step-down Dunnett test 299–301 step-down procedure 298

STMTS= statement, %GLIMMIX macro 330, 338

MIXPRINTALL option 330 OPTIONS= option 330

stratified Kappa statistic 365 stratified randomization 215, 225, 228

factorial stratification 227 strong learning algorithms 11

See also boosting Student’s two-sample t-test 122, 123–124 stumps

See recursive partitioning subject allocation

See allocation in randomized clinical trials SUBJECT option, RANDOM statement

(GLIMMIX) 292, 336 subject-specific models

See random-effects models for missing categorical data

subscales, identifying 379–382 subset algorithms 52 superiority testing (dose-response studies)

279–280 support vector machines 8

T

TABLES statement, FREQ procedure JT option 286

TABULATE procedure 101–102 Taves minimization algorithm 229, 232 TECHNIQUE= option, NLMIXED procedure

78–79 NEWRAP value 78 NRRIDG value 78 QUANEW value 78

test data See training and test sets

test-retest reliability 362, 372–376 therapeutic window 294 time stationarity of clearance 207–209 TOST assessment 199 total error, analytical procedures 85–88

profiles of 90–92

Page 455: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

442 Index

toxicology studies See nonclinical safety assessment

training and test sets 46 influential points in 48 leverage value 62–63 outliers in 48 selecting 47–51 solubility data 49–51 when observations are not in 61–63

TRAINTEST module 49–51 treatment group splitting, allocation for

221–224 TREATMENTS statement, PLAN procedure

101–102, 226 trend detection in dose-response studies

280–289 comparison of trend tests 287 contrast tests 281–284 isotonic tests 284–286 Jonckheere test 286–287 nonparametric tests 286

trend test (sequential testing method) 103–104 true value

See validation of analytical methods trueness of analytical procedures 85–88 TTEST procedure 122, 123–124 two-drug combination studies 153 two-factor ANOVA model, power evaluation in

102–106 two-parameter logistic models 160–161, 162,

164–165 two-sample experiments 118–129

power calculations 145–146 similar dispersion 121–123 unequal dispersion 123–129

two-sided dose-response studies 280 two-sided tests 245 two-tailed version of Wilcoxon Rank Sum test

122 Type I error rates

crucial 238, 239, 249–253, 260–263 dose-response studies 280 in sample-size analysis 238, 239, 241–243 TOST assessment 199 two-factor ANOVA models 106

Type II error rates crucial 238, 239, 249–253, 260–263 dose-response studies 280 sample-size analysis 238, 239, 241–243

TYPE= option RANDOM statement (GLIMMIX) 334, 336

REPEATED statement (%GLIMMIX) 330 MIXED procedure 203

U

U-shaped dose-response curves 304–306 ULOQ (upper limit of quantification) 92 umbrella-shaped dose-response curves 304–306 unequally weighted observations, boosting with

16–17 upper limit of quantification (ULOQ) 92 upper-tailed version of Wilcoxon Rank Sum test

122

V

validation cross-validation 22, 47 generalized boosting and 22 samples for 72, 94

validation of analytical methods 69–94 accuracy and precision 85–88 criteria for 73–74 decision rule 88–92 limits of 92–93 linearity 83–85 response function (calibration curve) 74–83 terminology 94

validity of outcome measures 361, 376–382 content validity 376 convergent and divergent 377 discriminant 377–378 responsiveness 376, 378

VAR statement NPAR1WAY procedure 121 REG procedure 58–59

variable block size, permuted block designs 217–219

variable ordering procedure 53–58 variable selection methods 51–58 Variable Selection node (SAS Enterprise Miner)

64–65 variance components structure (VC), one-factor

ANOVA model 110–112

W

weak learning algorithms 11–14 See also boosting

%WEEKINT macro 108 WEIGHT statement, MIXED procedure 75 _WEIGHT_ statement, NLIN procedure 77 weighted generalized estimating equations

(WGEE) 322–323, 329

Page 456: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Index 443

weighted Kappa statistic 365 WEIGHTS parameter (%OptimalDesign1) 161 WGEE (weighted generalized estimating

equations) 322–323, 329 WILCOXON option

EXACT statement (NPAR1WAY) 133 NPAR1WAY procedure 121, 133

Wilcoxon Rank Sum test 121, 124–125 lower-tailed version 122 rank-transformations and 129 two-tailed version 122 upper-tailed version 122

Williams trend test 284–285 sample size calculations 288

%WilliamsTest macro 285

Y

Y= option, GAM procedure 60 PARAM value 60 SPLINE value 60 SPLINE2 value 60

Page 457: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

444 Index

Page 458: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

Books Available from SAS Press

Advanced Log-Linear Models Using SAS®

by Daniel Zelterman

Analysis of Clinical Trials Using SAS®: A Practical Guideby Alex Dmitrienko, Geert Molenberghs, Walter Offen, and Christy Chuang-Stein

Annotate: Simply the Basicsby Art Carpenter

Applied Multivariate Statistics with SAS® Software,Second Editionby Ravindra Khattreeand Dayanand N. Naik

Applied Statistics and the SAS® Programming Language, Fifth Editionby Ronald P. Cody and Jeffrey K. Smith

An Array of Challenges — Test Your SAS® Skillsby Robert Virgile

Carpenter’s Complete Guide to the SAS® Macro Language, Second Editionby Art Carpenter

The Cartoon Guide to Statistics by Larry Gonick and Woollcott Smith

Categorical Data Analysis Using the SAS® System, Second Editionby Maura E. Stokes, Charles S. Davis, and Gary G. Koch

Cody’s Data Cleaning Techniques Using SAS® Softwareby Ron Cody

Common Statistical Methods for Clinical Research withSAS® Examples, Second Editionby Glenn A. Walker

The Complete Guide to SAS® Indexesby Michael A. Raithel

Data Management and Reporting Made Easy with SAS® Learning Edition 2.0by Sunil K. Gupta

Data Preparation for Analytics Using SAS®

by Gerhard Svolba

Debugging SAS® Programs: A Handbook of Tools and Techniquesby Michele M. Burlew

Decision Trees for Business Intelligence and Data Mining: Using SAS® Enterprise MinerTM

by Barry de Ville

Efficiency: Improving the Performance of Your SAS®

Applicationsby Robert Virgile

The Essential Guide to SAS® Dates and Timesby Derek P. Morgan

The Essential PROC SQL Handbook for SAS® Users

by Katherine Prairie

Fixed Effects Regression Methods for Longitudinal Data Using SAS®

by Paul D. Allison

Genetic Analysis of Complex Traits Using SAS®

Edited by Arnold M. Saxton

A Handbook of Statistical Analyses Using SAS®, Second Editionby B.S. Everittand G. Der

Health Care Data and SAS® by Marge Scerbo, Craig Dickstein,and Alan Wilson

The How-To Book for SAS/GRAPH® Softwareby Thomas Miron

In the Know ... SAS® Tips and Techniques FromAround the Globe, Second Editionby Phil Mason

Instant ODS: Style Templates for the Output Delivery Systemby Bernadette Johnson

Integrating Results through Meta-Analytic Review UsingSAS® Softwareby Morgan C. Wang and Brad J. Bushman

Introduction to Data Mining Using SAS® Enterprise MinerTM

by Patricia B. Cerrito

Learning SAS® in the Computer Lab, Second Editionby Rebecca J. Elliott

The Little SAS® Book: A Primerby Lora D. Delwiche and Susan J. Slaughter

The Little SAS® Book: A Primer, Second Editionby Lora D. Delwiche and Susan J. Slaughter (updated to include SAS 7 features)

support.sas.com/pubs

Page 459: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

The Little SAS® Book: A Primer, Third Editionby Lora D. Delwiche and Susan J. Slaughter(updated to include SAS 9.1 features)

The Little SAS® Book for Enterprise Guide® 3.0by Susan J. Slaughterand Lora D. Delwiche

The Little SAS® Book for Enterprise Guide® 4.1by Susan J. Slaughterand Lora D. Delwiche

Logistic Regression Using the SAS® System:Theory and Applicationby Paul D. Allison

Longitudinal Data and SAS®: A Programmer’s Guideby Ron Cody

Maps Made Easy Using SAS®

by Mike Zdeb

Models for Discrete Data by Daniel Zelterman

Multiple Comparisons and Multiple Tests Using SAS®

Text and Workbook Set(books in this set also sold separately)by Peter H. Westfall, Randall D. Tobias,Dror Rom, Russell D. Wolfinger,and Yosef Hochberg

Multiple-Plot Displays: Simplified with Macrosby Perry Watts

Multivariate Data Reduction and Discrimination with SAS® Softwareby Ravindra Khattree and Dayanand N. Naik

Output Delivery System: The Basicsby Lauren E. Haworth

Painless Windows: A Handbook for SAS® Users, Third Editionby Jodie Gilmore(updated to include SAS 8 and SAS 9.1 features)

Pharmaceutical Statistics Using SAS®: A Practical GuideEdited by Alex Dmitrienko, Christy Chuang-Stein, and Ralph D’Agostino

The Power of PROC FORMAT by Jonas V. Bilenas

PROC SQL: Beyond the Basics Using SAS®

by Kirk Paul Lafler

PROC TABULATE by Exampleby Lauren E. Haworth

Professional SAS® Programmer’s Pocket Reference, Fifth Editionby Rick Aster

Professional SAS® Programming Shortcuts, Second Editionby Rick Aster

Quick Results with SAS/GRAPH® Softwareby Arthur L. Carpenter and Charles E. Shipp

Quick Results with the Output Delivery Systemby Sunil K. Gupta

Reading External Data Files Using SAS®: Examples Handbookby Michele M. Burlew

Regression and ANOVA: An Integrated Approach Using SAS® Softwareby Keith E. Muller and Bethel A. Fetterman

SAS® for Forecasting Time Series, Second Editionby John C. Brocklebankand David A. Dickey

SAS® for Linear Models, Fourth Editionby Ramon C. Littell, Walter W. Stroup,and Rudolf J. Freund

SAS® for Mixed Models, Second Editionby Ramon C. Littell, George A. Milliken, Walter W. Stroup,Russell D. Wolfinger, and Oliver Schabenberger

SAS® for Monte Carlo Studies: A Guide for Quantitative Researchersby Xitao Fan, Ákos Felsovályi, Stephen A. Sivo,and Sean C. Keenan

SAS® Functions by Exampleby Ron Cody

SAS® Guide to Report Writing, Second Editionby Michele M. Burlew

SAS® Macro Programming Made Easy, Second Editionby Michele M. Burlew

SAS® Programming by Exampleby Ron Codyand Ray Pass

SAS® Programming for Researchers and Social Scientists, Second Editionby Paul E. Spector

SAS® Programming in the Pharmaceutical Industryby Jack Shostak

SAS® Survival Analysis Techniques for Medical Research, Second Editionby Alan B. Cantor

SAS® System for Elementary Statistical Analysis,Second Editionby Sandra D. Schlotzhauerand Ramon C. Littell

SAS® System for Regression, Third Editionby Rudolf J. Freund and Ramon C. Littell

SAS® System for Statistical Graphics, First Editionby Michael Friendly

˝

support.sas.com/pubs

Page 460: Praise from the Experts - bayanbox.irbayanbox.ir/.../Pharmaceutical-Statistics-Using-SAS.pdf · Praise from the Experts “Pharmaceutical Statistics Using SAS contains applications

The SAS® Workbook and Solutions Set(books in this set also sold separately)by Ron Cody

Selecting Statistical Techniques for Social Science Data:A Guide for SAS® Usersby Frank M. Andrews, Laura Klem, Patrick M. O’Malley,Willard L. Rodgers, Kathleen B. Welch,and Terrence N. Davidson

Statistical Quality Control Using the SAS® Systemby Dennis W. King

A Step-by-Step Approach to Using the SAS® Systemfor Factor Analysis and Structural Equation Modelingby Larry Hatcher

A Step-by-Step Approach to Using SAS® for Univariate and Multivariate Statistics, Second Editionby Norm O’Rourke, Larry Hatcher,and Edward J. Stepanski

Step-by-Step Basic Statistics Using SAS®: Student Guide and Exercises(books in this set also sold separately)by Larry Hatcher

Survival Analysis Using SAS®:A Practical Guideby Paul D. Allison

Tuning SAS® Applications in the OS/390 and z/OS Environments, Second Editionby Michael A. Raithel

Univariate and Multivariate General Linear Models:Theory and Applications Using SAS® Softwareby Neil H. Timmand Tammy A. Mieczkowski

Using SAS® in Financial Researchby Ekkehart Boehmer, John Paul Broussard,and Juha-Pekka Kallunki

Using the SAS® Windowing Environment: A Quick Tutorialby Larry Hatcher

Visualizing Categorical Databy Michael Friendly

Web Development with SAS® by Example, Second Editionby Frederick E. Pratter

Your Guide to Survey Research Using the SAS® Systemby Archer Gravely

JMP® Books

JMP® for Basic Univariate and Multivariate Statistics: A Step-by-Step Guideby Ann Lehman, Norm O’Rourke, Larry Hatcher,and Edward J. Stepanski

JMP® Start Statistics, Third Editionby John Sall, Ann Lehman,and Lee Creighton

Regression Using JMP® by Rudolf J. Freund, Ramon C. LIttell,and Lee Creighton

support.sas.com/pubs